EP2431386B1 - Na+/Mg2+ exchanger - Google Patents

Na+/Mg2+ exchanger Download PDF

Info

Publication number
EP2431386B1
EP2431386B1 EP10177510.4A EP10177510A EP2431386B1 EP 2431386 B1 EP2431386 B1 EP 2431386B1 EP 10177510 A EP10177510 A EP 10177510A EP 2431386 B1 EP2431386 B1 EP 2431386B1
Authority
EP
European Patent Office
Prior art keywords
protein
slc41a1
cells
seq
homo sapiens
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
EP10177510.4A
Other languages
German (de)
French (fr)
Other versions
EP2431386A1 (en
Inventor
Monika Röntgen
Martin Kolisek
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Fbn - Leibniz-Institut fur Nutztierbiologie
Original Assignee
Fbn - Leibniz-Institut fur Nutztierbiologie
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Fbn - Leibniz-Institut fur Nutztierbiologie filed Critical Fbn - Leibniz-Institut fur Nutztierbiologie
Priority to EP10177510.4A priority Critical patent/EP2431386B1/en
Priority to PCT/EP2011/065979 priority patent/WO2012035088A1/en
Priority to JP2013528667A priority patent/JP2013543486A/en
Priority to CA2811538A priority patent/CA2811538C/en
Priority to US13/823,383 priority patent/US20130174289A1/en
Publication of EP2431386A1 publication Critical patent/EP2431386A1/en
Application granted granted Critical
Publication of EP2431386B1 publication Critical patent/EP2431386B1/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K67/00Rearing or breeding animals, not otherwise provided for; New breeds of animals
    • A01K67/027New breeds of vertebrates
    • A01K67/0275Genetically modified vertebrates, e.g. transgenic
    • A01K67/0276Knockout animals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/10Processes for the isolation, preparation or purification of DNA or RNA
    • C12N15/1034Isolating an individual clone by screening libraries
    • C12N15/1079Screening libraries by altering the phenotype or phenotypic trait of the host
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6872Intracellular protein regulatory factors and their receptors, e.g. including ion channels
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/07Animals genetically altered by homologous recombination
    • A01K2217/075Animals genetically altered by homologous recombination inducing loss of function, i.e. knock out
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/20Animal model comprising regulated expression system
    • A01K2217/203Animal model comprising inducible/conditional expression system, e.g. hormones, tet
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2267/00Animals characterised by purpose
    • A01K2267/03Animal model, e.g. for test or diseases
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value

Definitions

  • the magnesium ion Mg 2+ is a vital element essentially involved in numerous physiological processes at the cell, organ and organism level [Kolisek et al., 2007]. Therefore, the biologically active free intracellular Mg 2+ concentration ([Mg 2+ ] i ) has to be adjusted to its optimal level of 0.5 to 1.2 mM. In this process transmembrane extrusion of Mg 2+ has been shown to be critically involved (Schweigel et al., 2006).
  • Na + -dependent efflux of intracellular Mg 2+ has been shown to operate in the majority of examined cells including ventricular myocytes, vascular smooth muscle cells, hepatocytes, thymocytes, neurons, renal and gastrointestinal epithelial cells (Kolisek et al., 2007; Schweigel et al., 2006).
  • the Na + /Mg 2+ exchanger is also a candidate mechanism for the extrusion of Mg 2+ across the basolateral membrane of the Mg 2+ absorbing and reabsorbing intestinal and renal epithelia (Quamme, 1989) making it important for control of body Mg 2+ homeostasis (balance).
  • Mg 2+ transporters found by a differential gene expression approach (Goytain and Quamme, 2005a-d, Goytain et al., 2007), are MagT1 (Magnesium Transporter 1), ACDP 2 (Ancient conserveed Domain Protein; subtype 2), the protein NIPA1 (Nonimprinted in Prader-Willi/Angelman), SLC41A1 and SLC41A2 (Solute Carrier family 41; subfamily A; members 1 and 2).
  • WO 0190360 discloses human SLC41A1.
  • mice (m)SLC41A1, mSLC41A2, mACDP 2 and mMagT1 all as being Mg 2+ channels mediating influx of the ion.
  • SLC41A1 has been shown to have partial sequence homology [Wabakken et al., 2003] to a specific region in the transmembrane domain of the bacterial Mg 2+ transport system MgtE described by Smith et al. (1995). It is interesting to note, that the MgtE proteins have been discovered by complementation assays and that they show no homology to other groups of bacterial Mg 2+ transporters operating as pumps (MgtA, MgtB) or channels (CorA).
  • SLC41A1 transcripts have been found to be highly abundant in heart, brain, kidney, liver, muscle, testis, thyroid gland, lymphocytes and to a lesser extent in most other tissues including colon and small intestine. Neither the functions of MgtE nor of SLC41A1 have been thoroughly characterized.
  • the object of the present invention was the identification of the encoding gene(s) and the salvation of the protein structure of the Na + /Mg 2+ exchanger.
  • the object was solved by the finding that the protein SLC41A1 is the cellular Na + /Mg 2+ exchanger. It could be proven that the protein SLC41A1 mediates Mg 2+ extrusion and its mode of operation could be clarified.
  • the naming SEQ ID NO 1 is used for the protein SLC41A1 (GenBank Accession No. NP_776253) and the naming SEQ ID NO 2 is used for the underlying mRNA sequence of SLC41A1 (GenBank Accession No. NM_173854).
  • the cDNA sequence is represented in SEQ ID NO 3.
  • SEQ ID NO 4 represents the SLC41A1 from mouse (Mus musculus, complete cds, GenBank Accession No. BC116280.1, CDS: 802>2340) and SEQ I D NO 5 is the related protein sequence (Mus musculus, NCBI Reference Sequence: NP_776290.1).
  • the invention relates to methods to identify new SLC41A1 functions at the cell, tissue, organ and organism level. In part, it is related to methods useful in (a) identifying molecules that bind SLC41A1 polypeptides, which (b) modulate SLC41A1 related Na + /Mg 2+ exchanger activity or its cellular or tissue specific expression.
  • the invention comprises SLC41A1 mutation libraries. Also disclosed are SLC41A1 specific antibodies and their generation as well as an inducible conditional knock out mice model and a conditional inducible knock out cell line.
  • the SLC41A1 mutation libraries comprise at least one of the following mutation clusters i, ii, iii, iv of the amino acid sequence of SEQ ID NO 1 (indicated in brackets are the triplet changes of the cDNA of SEQ ID NO 3):
  • the mutations described in cluster i) are believed to be useful for the identification of SLC41A1 variants showing a modified phosphorylation pattern and therefore, an impaired or modified function of the carrier.
  • the LZ domain is known to be critical in dimerization and thus, for the complex forming ability of SLC41A1 which is essential for its normal function as a carrier (Kolisek et al., 2008). Mutations in the LZ domain (cluster iii) are believed to be helpful in the generation of SLC41A1 variants with changed transport and regulatory characteristics and/or mode of action.
  • Each of these sequences is combined in a mutation library, which comprises at least all sequences of one of the mutation clusters i) to iv).
  • the most preferred embodiment is a mutation library, which combines all four mutation clusters i) to iv).
  • the expected outcome of mutation clusters i, ii and iv is also a definitive confirmation of the SLC41A1 orientation in cytoplasmatic membrane of the cell. As shown by Kolisek et al. (2008), a predicted model which comprises 10 TMs and both N- and C- terminus oriented intracellularly is most likely.
  • Respective mutated SLC41A1 inserts are cloned into cloning vector pUC19, the selected clones are controlled for the proper insert orientation and sequenced for verification purposes.
  • the constructed library is subcloned into respective mammalian expression vector(s) and used for functional screening in an inducible conditional SLC41A1 knockout cell line or into cells of a transgenic conditional knockout animal, which both will be described in more detail below.
  • a protein consisting of an amino acid sequence SEQ ID NO 1 or a protein fragment thereof, in which one or more amino acids are substituted, deleted or added and which has an identity on the amino acid level with SEQ ID NO 1 of at least 90%, preferably at least 95%, more preferably 98%, for the generation and identification of specific antibodies against the cellular Na + /Mg 2+ exchanger.
  • identity of at least 90% always means that this comprises preferably an identity of at least 95%, more preferably an identity of 98%, regardless whether proteins or DNAs are concerned.
  • the related method of identifying antibodies against the cellular Na + /Mg 2+ exchanger comprises the steps of:
  • the complex is preferably detected by a secondary antibody.
  • the primary und/or the secondary antibody comprise(s) a label, which is selected from the group of radioactive label, fluorescent label, enzymatic label, or biotin label.
  • kits for identifying antibodies against the Na + /Mg 2+ exchanger comprising a protein consisting of an amino acid sequence of SEQ ID NO 1 or a protein fragment thereof, in which one or more amino acids are substituted, deleted or added and which has an identity on the amino acid level with SEQ ID NO 1 of at least 90%.
  • a protein consisting of an amino acid sequence of SEQ ID NO 1 or a protein fragment thereof, in which one or more amino acids are substituted, deleted or added and which has an identity on the amino acid level with SEQ ID NO 1 of at least 90% in a method which uses the protein in order to generate and produce antibodies, preferably monoclonal antibodies, against the Na + /Mg 2+ exchanger.
  • These antibodies can be used for the detection of the presence, absence and or amount and for the interaction with the cellular Na + /Mg 2+ exchanger, which has a protein having the amino acid sequence of SEQ ID NO 1 or a protein fragment thereof, in which one or more amino acids are substituted, deleted or added and which has an identity on the amino acid level with SEQ ID NO 1 of at least 90%.
  • the antibodies may be polyclonal or monoclonal but monoclonal antibodies are preferred.
  • a protein consisting of an amino acid sequence of SEQ ID NO 1 or a protein fragment thereof, in which one or more amino acids are substituted, deleted or added and which has an identity on the amino acid level with SEQ ID NO 1 of at least 90% for the identification of specific protein binding partners that can form stable or transient protein complexes with the cellular Na + /Mg 2+ exchanger.
  • the related method of identifying specific protein complex partners for the Na + /Mg 2+ exchanger comprises the steps of:
  • kits for identifying specific protein binding partners forming stable or transient protein complexes with the Na + /Mg 2+ exchanger comprising a protein having an amino acid sequence of SEQ ID NO 1 or a protein fragment thereof, in which one or more amino acids are substituted, deleted or added and which has an identity on the amino acid level with SEQ ID NO 1 of at least 90%.
  • Especially the substances listed below prove suitable as protein complex partner for the Na + /Mg 2+ exchanger, which is identical to a protein having an amino acid sequence of SEQ ID NO 1 or a protein fragment thereof, in which one or more amino acids are substituted, deleted or added and which has an identity on the amino acid level with SEQ ID NO 1 of at least 90%:
  • SLC41A1 complex-forming partners were screened out by the split-ubiquitin method, which is well known in the art and are listed in detail in Table 1.
  • the ubiquitin-based split protein sensor (USPS) is the method that makes it possible to detect and monitor protein-protein interactions as a function of time, at the natural sites of these interactions in a living cell. Details on the method can be found in Johnsson and Varshavsky, 1994.
  • 3-beta-hydroxysteroid-Delta(8),Delta(7)-isomerase an enzyme which catalyzes the conversion of Delta(8)-sterols to their corresponding Delta(7)-isomers.
  • 3-beta-hydroxysteroid-Delta(8),Delta(7)-isomerase interacted with SLC41A1.
  • Defects in 3-beta-hydroxysteroid-Delta(8),Delta(7)-isomerase are the cause of chondrodysplasia punctata also known as Conradi-Hunermann-Happle syndrome (Derry et al, 1999).
  • the third strongest SLC41A1-binding candidates were Peptidyl-prolyl cis-trans isomerase and UPF0480 protein C15orf24.
  • One interesting candidate is the Kunitz type protease inhibitor 2. When unfunctional, the protein cause a rare, inherited diarrhea of infancy named congenital secretory sodium diarrhea type 3 (DIAR3) or congenital sodium diarrhea (CSD).
  • modulators which either increase or decrease the SLC41A1-related Na + /Mg 2+ exchanger activity and are thus called modulators.
  • modulators modulate SLC41A1-related Na + /Mg 2+ exchanger activity either by direct interaction with structural components of the SLC41A1 molecule or by the induction of cellular signal cascades resulting in modifications of the SLC41A1 protein. Therefore, some of these modulators are hormones or elements of cellular signal cascades.
  • Main sites for posttranslational modifications and thus, regulation/modulation of SLC41A1-related Na + /Mg 2+ exchanger activity were the following:
  • imipramine, quinidin, cyclic adenosine monophosphate (cAMP) analogues, ⁇ -adrenergic and ⁇ 1 -adrenergic agonists and the hormones angiotensin II and insulin showed strong modulating effects. It could be demonstrated that replacement of extracellular Na + with N-methyl-D-glutamine (NMDG) or application of the unspecific Na + /Mg 2+ exchanger inhibitors imipramine and quinidin strongly decrease SLC41A1-mediated efflux.
  • NMDG N-methyl-D-glutamine
  • the above mentioned modulators especially imipramine, quinidin, DIDS, lidocaine, nifedipine, benzamil, KB-R7943, SEA0400, amiloride, agonists or antagonists of receptors for glucocorticoids, aldosterone, angiotensin II, insulin, glucagon, PGE 2 and catecholamines ( ⁇ -adrenergic and/or ⁇ 1-adrenergic agents), cAMP or cGMP analogues, taurine, genistein, calphostin C, phorbol 12,13-dibutyrate, L-NMMA, NOR1, ODQ, forskolin, indomethacin, arachidonic acid, spermine, sper
  • the above-mentioned antibodies, protein binding partners and the above named modulators or their analogues or agents acting as agonists or antagonists of these modulators are believed to be useful for treating patients having conditions that are characterizid by increased or decreased Mg 2+ efflux via their Na + /Mg 2+ exchanger expressing cells.
  • Such conditions of disturbed or dysregulated Na + /Mg 2+ exchanger activity have been described in various disease complexes such as cardiovascular disorders, metabolic and endocrine dysfunctions, neurologic diseases, cancer, pre-eclampsia, eclampsia, fetal growth retardation and premature labor.
  • cardiovascular diseases are listed from top to bottom according to the present evidence that their pathogenesis is related to an increased or decreased Mg 2+ efflux via Na + /Mg 2+ exchanger expressing cells.
  • the strength of evidence is indicated by stars in the last column.
  • Cardiovascular diseases have the strongest evidence that they are related to the Na + /Mg 2+ exchanger.
  • the treatment of cardiovascular diseases is the most relevant application for the present invention, i.e. for the antibodies, the special protein binding partners as outlined above or for the specific modulators outlined above.
  • the nucleotide sequence of SEQ ID NO 3 (cDNA) is of special use for the gene therapy of these diseases, i.e. for the treatment of a disease from the group of cardiovascular diseases, metabolic or endocrine dysfunctions, neurological diseases, cancer, pre-eclampsia, eclampsia, fetal growth retardation and premature labor, preferably for the treatment of hypertension, ischemic heart disease, myocardial infarction, cardiac dysfunction in relation to diabetes mellitus, pre-eclampsia, eclampsia, diabetes mellitus (type I), non-insulin-dependent diabetes, mukoviszidose, cystic fibrosis (type II), parkinson disease, morbus parkinson, stroke, prostate cancer as well as aggressive behaviour or high stress sensitivity.
  • a model based on a non-human mammal preferably a mouse, having an inducible conditional system leading to a functional knock out of SLC41A1, i.e. a genetically modified conditional knock out non-human mammal, which is characterized in that the SCL41A1 gene is inducible conditionally knockable (knock out), i.e. it can be knocked-out in a time (e.g., at embryonic, young or adult stage of development) or tissue-specific (particular organ or cell type) manner.
  • the mouse is referred to because this is the preferred non-human mammal.
  • the mouse model was developed on the basis of the gene sequence of SLC41A1 (SEQ ID NO 3, GenBank Accession No. NM_173854).
  • a SLC41A1 inducible conditional KO mouse was developed by homologous recombination using 129 embryonic stem cells. This mouse line was then crossed with Cre recombinase (CR) or ligand-dependent Cre recombinase (LCR) expressing mice for the generation of tissue specific or time dependent targeted inactivation of murine SLC41A1.
  • CR Cre recombinase
  • LCR ligand-dependent Cre recombinase
  • LCR are fusion proteins between Cre and the ligand-binding domains (LBDs) of steroid receptors.
  • LBD ligand-binding domains
  • the LBD has been mutated so that it does not respond to its natural ligand yet binds a synthetic ligand.
  • tamoxifen-activated CreERT recombinase modified pacman
  • ER mutated estrogen receptor
  • OHT 4-hydroxytamoxifen
  • the recombinase is located in the cytoplasm.
  • Binding of OHT to the LBD results in the translocation of the recombinase into the nucleus where it can recombine its IoxP substrates (Feil R., 2007).
  • the LCR system is widely used for the generation of inducible conditional KO mice in experiments with time dependent settings but also in those with demand for tissue specificity.
  • Mouse lines expressing CreERT2 recombinase in tissue specific manner are listed below and the newly developed conditional KO SLC41A1 mouse can be bread with one of the available CreERT2 mouse lines in dependence of specific experimental needs.
  • mice expressing the CreERT2 recombinase in soecific tissues:
  • the transgenic conditional knock out mouse is characterized in that the genome comprises a construct, which allows conditional knock out of the SCL41A1.
  • construct refers to a polynucleotide, which was exogenously inserted into the genom of the animal.
  • the animal can be heterozygous or homozygous but is preferably homozygous with respect to the knock out gene ( -/- ).
  • the construct comprises the SCL41A1 gene of SEQ ID NO 4 (murine gene) flanked by two IoxP DNA sequence elements (IoxP sites), i.e. they are preferably situated 3' and 5' of the SCL41A1 gene.
  • transgenic inducibleconditional knock out cell or cell line a transgenic inducibleconditional knock out cell or cell line.
  • Cells can be taken from the transgenic inducible conditional knock out non-human mammal or can originate from separate cell lines.
  • primary cell cultures from the heart (cardiomyocytes), from the vasculature (e.g. aorta), from the liver (hepatocytes) or from the brain or CNS/microglial cells of these animals.
  • stable human cell lines more preferably stable human embryonic kidney cell line (ACC-305/DSMZ) with inducible conditionally knocked out SLC41A1 gene, HEK293 (hSLC41A1 -/- ) or stable human B-cell precursor leukemia cell line (ACC-128/DSMZ) with inducible conditionally knocked out SLC41A1 gene, NALM-6 (hSLC41A1 -/- ).
  • stable human embryonic kidney cell line ACC-305/DSMZ
  • HEK293 hSLC41A1 -/-
  • stable human B-cell precursor leukemia cell line ACC-128/DSMZ
  • NALM-6 hSLC41A1 -/-
  • Cell lines will be generated according to the technique described by Shibahara Kei-ichi et al..
  • Mammalian vectors which comprise mutated forms of human SLC41A1 are transferred or electroporated into these primocultured cells of or into cells of the stable human cell lines, resulting in cells having in their genome mammalian vectors with a mutated human SLC41A1.
  • the mutated forms of human SLC41A1 are as described above with respect to the mutation libraries, i.e. each of these mutated sequences can be present in the vector.
  • HEK293-(FLAG-SLC41A1) cell line growth media and culture conditions
  • Non-induced (-tet) and induced (+tet, 15-18 h) HEK293-(FLAG-SLC41A1) cells (107 cells ml -1 ) were lysed for 30 min at 4 °C in Tris buffer (pH 7.5) containing 1% Triton X-100 (Bio-Rad) and protease inhibitors (Perbio-ThermoFisher).
  • the uninduced (-tet) and induced (+tet) HEK293-(SLC41A1) cells were rinsed twice with ice-cold, completely divalent-free Dulbecco's phosphate-buffered saline (DPBS), detached by use of Hytase (Perbio Science, Bonn, Germany), centrifuged, washed twice in completely Ca 2+ - and Mg 2+ -free Hank's balanced solution (CMF-HBS, PAN Biotech, Aidenbach, Germany) complemented with 10 mmol.l -1 HEPES and 1.36 mmol.l -1 L-glutamine (CMF-HBS+) and finally re-suspended in the same solution.
  • DPBS Dulbecco's phosphate-buffered saline
  • the latter was determined as the intracellular Mg 2+ concentration ([Mg 2+ ] i ) decrease over a 20-min period.
  • -tet and +tet HEK293-(SLC41A1) were Mg-loaded by a 20-min incubation in CMF-HBS+ containing 0, 2, 5 or 10 mmol.l -1 Mg.
  • An extracellular Mg 2+ concentration ([Mg 2+ ] e ) of 10 mmol.l -1 was selected for all further experiments. After loading, the remaining extracellular Mg 2+ was removed by washing the cells twice in CMF-HBS+ and, after re-suspending the cells in CMF-HBS+, measurements were started.
  • the final [Mg 2+ ] i was determined as the mean [Mg 2+ ] i of the last 10 seconds of the measurement. Thus, for the calculation of any given [Mg 2+ ] i , 500 data points were used. If not otherwise stated, data are presented as means ⁇ standard error (SE).
  • Table 5 List of special media Name Remarks Producer -Cl - , -Mg 2+ , -Ca 2+ Hank's medium Cl - , Mg 2+ , Ca 2+ free medium (Cl - has been substituted with glutamate) Completely Mg 2+ - and Na + -free PBS Without Ca 2+ / Mg 2+ , Na + substituted by N-methyl-D-glucamine (NMDG) No. LZ 1825, Biochrom, Berlin, Germany Ca 2+ - and Mg 2+ -free medium 199 Hanks Without Ca 2+ / Mg 2+ and phenol red No.
  • Mg 2 + efflux from hSLC41A1 depends on extracellular Na + and is inhibited by the unspecific Na + / Mg 2 + exchanger inhibitors imipramine and quinidine.
  • Mg-loaded (10 mM) -tet and +tet HEK293-(SLC41A1) cells were suspended in absolutely Mg-free media containing 145 mM Na + or in the same medium but all Na + iso-osmotically replaced by the organic non-permeant cation NMDG and the [Mg 2+ ] i was measured over a 20-minute period ( figure 2A ).
  • a steady [Mg 2+ ] i of 0.45 ⁇ 0.03 mM was determined.
  • Mg 2+ -loaded (20 min, 10 mM) +tet HEK293-(SLC41A1) cells were incubated in a completely Mg 2+ -free medium with an [Na + ] e of 145 mM without or with 250 ⁇ M imipramine ( Fig. 3A ) or 100 ⁇ M quinidin ( Fig. 3C ), respectively, and the [Mg 2+ ] i was measured for 20 min. To correct for molecular leakiness, control measurements were done with -tet cells.
  • FIG. 3A shows that application of imipramine diminished the SLC41A1-related Mg 2+ efflux.
  • Data are summarized in figure 3B , which shows that inhibitor application reduced Mg 2+ efflux from 341 ⁇ 35 to 129 ⁇ 32 ⁇ M/20min corresponding to 64 ⁇ 9%.
  • Figure 3C shows a summary of the inhibitory effect of 100 ⁇ M quinidin on SLC41A1-mediated Mg 2+ efflux. It decreased from 327 ⁇ 71 ⁇ M/20 min to 176 ⁇ 0.5 ⁇ M/20 min corresponding to a 38 ⁇ 7% reduction.
  • hSLC41A1-mediated Mg 2+ efflux is stimulated by dB-cAMP and angiotensin II but inhibited by insulin.
  • angiotensin II stimulated SLC41A1-related Mg 2+ efflux by 19,5%.
  • insulin reduced the SLC41A1-mediated Mg 2+ efflux by 38% (200 ⁇ U7ml) and 68% (400 ⁇ U/ml), respectively.
  • the Ca 2 + channel inhibitor Verapamil has no effect on hSLC41A1-mediated Mg 2+ efflux.

Description

  • The magnesium ion Mg2+ is a vital element essentially involved in numerous physiological processes at the cell, organ and organism level [Kolisek et al., 2007]. Therefore, the biologically active free intracellular Mg2+ concentration ([Mg2+]i) has to be adjusted to its optimal level of 0.5 to 1.2 mM. In this process transmembrane extrusion of Mg2+ has been shown to be critically involved (Schweigel et al., 2006). So far, there is no evidence for the existence of an outwardly oriented Mg-ATPase, however, by functional studies two main efflux systems, namely an anion-dependent mechanism [Ödblom and Handy, 1999] and a putative Na+/Mg2+ exchanger [Schweigel et al., 2006, 2009], have been proposed. After its first description in chicken and turkey red blood cells (Günther et al., 1984), Na+-dependent efflux of intracellular Mg2+ has been shown to operate in the majority of examined cells including ventricular myocytes, vascular smooth muscle cells, hepatocytes, thymocytes, neurons, renal and gastrointestinal epithelial cells (Kolisek et al., 2007; Schweigel et al., 2006). The Na+/Mg2+ exchanger is also a candidate mechanism for the extrusion of Mg2+ across the basolateral membrane of the Mg2+ absorbing and reabsorbing intestinal and renal epithelia (Quamme, 1989) making it important for control of body Mg2+ homeostasis (balance). The near-ubiquity of the Na+/Mg2+ exchanger attests to its functional importance, as is substantiated by its participation in the pathogenesis of a growing collection of human diseases (Kimura, 2007). Disturbance or dysregulation of the antiporter seem to be involved in primary hypertension, ischemic heart disease, eclampsia, diabetes and cystic fibrosis to name a few (Kisters et al., 1999; Vormann et al., 1992; Kimura, 2007; Kumeda and Inaba, 2007).
  • All approaches used so far, in order to identify the encoding gene(s) and/or to solve the protein structure of the Na+/Mg2+ exchanger, e.g. establishment of a high (> 100 mM)-Mg2+ resistant cell line (Watanabe et al., 2005), failed. Only the protein structure of the Na+/Mg2+ exchanger will allow for a detailed investigation of the modalities of its operation and regulation as well as for the development of specific inhibitors or agonists.
  • Within the last years several candidate genes have been characterized encoding for proteins involved in the regulation of eukaryotic cells Mg2+ homeostasis (summarized in [Sponder et al., 2010]). Only few of them have been clearly shown to be directly and specifically involved in Mg2+ transport processes. The best characterized are the TRPM6 and TRPM7 channels of the Melastatin-related Transient Receptor Potential family considered as being the major Mg2+ influx pathways of mammalian cells and the mitochondrial channel MRS2 (Mitochondrial RNA Splicing member 2) that mediates Mg2+ influx into the mitochondrial matrix [Schmitz, 2003; Kolisek, 2003, Schlingmann et al., 2002]. Other putative Mg2+ transporters, found by a differential gene expression approach (Goytain and Quamme, 2005a-d, Goytain et al., 2007), are MagT1 (Magnesium Transporter 1), ACDP 2 (Ancient Conserved Domain Protein; subtype 2), the protein NIPA1 (Nonimprinted in Prader-Willi/Angelman), SLC41A1 and SLC41A2 (Solute Carrier family 41; subfamily A; members 1 and 2).
  • WO 0190360 discloses human SLC41A1.
  • When over-expressed in Xenopus laevis oocytes, a genetically heterologous system, the group of Quamme [Goytain and Quamme, 2005a-d; Goytain et al., 2007] characterized mouse (m)SLC41A1, mSLC41A2, mACDP 2 and mMagT1 all as being Mg2+ channels mediating influx of the ion. However, with the exception of MagT1 the transport was not very specific to Mg2+ and, when over-expressed in human-derived cells hSLC41A2 [Sahni et al., 2007], hMagT1 [Zhou and Clapham, 2009] and hSLC41A1 (Kolisek et al., 2008) all failed to induce Mg2+ evoked currents. Moreover, by using tet-induced over-expression of hSLC41A1 in HEK293 cells strong evidence was found for human SLC41A1 as being a Mg2+ carrier localized to the cytoplasmatic membrane and mediating efflux of the ion [Kolisek et al., 2008]. SLC41A1 has been shown to have partial sequence homology [Wabakken et al., 2003] to a specific region in the transmembrane domain of the bacterial Mg2+ transport system MgtE described by Smith et al. (1995). It is interesting to note, that the MgtE proteins have been discovered by complementation assays and that they show no homology to other groups of bacterial Mg2+ transporters operating as pumps (MgtA, MgtB) or channels (CorA). Human [Wabbaken et al., 2003] and mice [Goytain and Quamme, 2005c] SLC41A1 transcripts have been found to be highly abundant in heart, brain, kidney, liver, muscle, testis, thyroid gland, lymphocytes and to a lesser extent in most other tissues including colon and small intestine. Neither the functions of MgtE nor of SLC41A1 have been thoroughly characterized.
  • Thus, the object of the present invention was the identification of the encoding gene(s) and the salvation of the protein structure of the Na+/Mg2+ exchanger.
  • The object was solved by the finding that the protein SLC41A1 is the cellular Na+/Mg2+ exchanger. It could be proven that the protein SLC41A1 mediates Mg2+ extrusion and its mode of operation could be clarified. In the following, the naming SEQ ID NO 1 is used for the protein SLC41A1 (GenBank Accession No. NP_776253) and the naming SEQ ID NO 2 is used for the underlying mRNA sequence of SLC41A1 (GenBank Accession No. NM_173854). The cDNA sequence is represented in SEQ ID NO 3. SEQ ID NO 4 represents the SLC41A1 from mouse (Mus musculus, complete cds, GenBank Accession No. BC116280.1, CDS: 802>2340) and SEQ I D NO 5 is the related protein sequence (Mus musculus, NCBI Reference Sequence: NP_776290.1).
  • The invention relates to methods to identify new SLC41A1 functions at the cell, tissue, organ and organism level. In part, it is related to methods useful in (a) identifying molecules that bind SLC41A1 polypeptides, which (b) modulate SLC41A1 related Na+/Mg2+ exchanger activity or its cellular or tissue specific expression.
  • Thus, the invention comprises SLC41A1 mutation libraries. Also disclosed are SLC41A1 specific antibodies and their generation as well as an inducible conditional knock out mice model and a conditional inducible knock out cell line.
  • The SLC41A1 mutation libraries comprise at least one of the following mutation clusters i, ii, iii, iv of the amino acid sequence of SEQ ID NO 1 (indicated in brackets are the triplet changes of the cDNA of SEQ ID NO 3):
    1. i) One mutation library comprises site-directed mutants of SEQ ID NO 1, wherein a single amino acid (aa) is substituted in position: 21 S, 25S, 73S, 76S, 80S, 89S, 97S, 158S, 309S, 394S, 408S, 81T, 168T, 509T and 278Y, respectively.
      In other words, a single aa serine (S), threonine (T) or tyrosine (Y) is replaced at one of the following positions of SEQ ID NO 1 by another aa:
      21 S>N (TCT>AAT), 25 S>Q (TCA>CAA), 73 S>N (AGC>AAC), 76 S>N (AGT>AAT), 80 S>N (AGC>AAC), 89 S>N (TCC>AAC), 97 S>N (TCC>AAC), 158 S>N (TCC>AAC), 309 S>N (AGT>AAT), 394 S>N (AGT>AAT) and 408 S>Q (TCA>CAA), 81 T>Q (ACA>CAA), 168 T>Q (ACA>CAA), 509 T>Q (ACG>CAG) and 278 Y>N (TAC>AAC), respectively. Comprised are also combinations of these mutations, i.e. at least one mutation of SEQ ID NO 1, wherein a plurality (2-15) of these substitutions is present. Preferably, 2-5 aa's are replaced, most preferably 2 or 3. In other words, the mutation library of cluster i) consists of 15 peptides with single aa mutation and at least one mutation of SEQ ID NO 1, wherein a plurality (2-15) of these substitutions is present.
    2. ii) Another mutation library comprises mutations of the transmembrane domains (TM) and N-, C- termini mutants of SEQ ID NO 1:
      Based on SEQ ID NO 1, respective deletions (Δ) of the amino acid sequences were present in:
      1. a) N-terminus and C-terminus flanking region: Δaa 2-30, Δaa 31-60, Δaa 61-90 and Δaa 2-60, Δaa 2-90, Δaa 504-513, respectively (6 mutated proteins) and
      2. b) membrane spanning domains of SEQ ID NO 1 of highly probable TM10 model (TMP): Δaa 96-205 (TM1+TM2); Δaa 225-279 (TM3+TM4); Δaa 286-337(TM5+TM6); Δaa 348-427 (TM7+TM8) and Δaa 439-503 (TM9+TM10), respectively (5 mutated proteins)
        and
      3. c) membrane spanning domains of less-probable TM11 model (SOSUI): Δaa 99-150 (TM1+TM2); Δaa 180-241 (TM3+TM4); Δaa 256-306 (TM5+TM6); Δaa 317-368 (TM7+TM8); Δaa 408-459 (TM9+TM10) and Δaa 437-503 (TM10+TM11), respectively (6 mutated proteins).
        Overall, cluster ii) comprises 17 mutated proteins.
    3. iii) Another mutation library comprises leucine zipper (LZ) mutants of SEQ ID NO 1:
      These SLC41A1 mutants comprise a deletion (Δ) respective to SEQ ID NO 1 of
      1. a) Δaa 482-503 and
      2. b) aa substitutions in LZ domain in positions 482 L>V (TTG>GTG), 489 L>V (CTT>GTT), 494 L>V (CTA>GTA), 496 L>V (CTC>GTC), 501 L>V (CTC>GTC) and 503 L>V (CTC>GTC), respectively (7 mutated proteins).
    4. iv) Another mutation library comprises external loops (EL) mutants of SEQ ID NO 1:
      These SLC41A1 mutants comprise deletions (Δ) respective to SEQ ID NO 1 of the amino acid sequences in EL: Δaa 128-179, Δaa 247-254, Δaa 310-315, Δaa 369-405 and Δaa 468-478 respectively (5 mutated proteins).
  • The mutations described in cluster i) are believed to be useful for the identification of SLC41A1 variants showing a modified phosphorylation pattern and therefore, an impaired or modified function of the carrier.
  • Mutations in the membrane spanning domains (cluster ii) and EL mutations (cluster iv) are believed to be useful for the identification of the minimal sequential configuration necessary to maintain the Mg2+/Na+ exchange (efflux) function of SLC41A1 in vivo and of the Mg2+ binding motif(s) / the Mg2+ binding site. Thus, they will be helpful to generate more active SLC41A1 variants or SLC41A1 variants with impaired function of the SLC41A1 carrier.
  • The LZ domain is known to be critical in dimerization and thus, for the complex forming ability of SLC41A1 which is essential for its normal function as a carrier (Kolisek et al., 2008). Mutations in the LZ domain (cluster iii) are believed to be helpful in the generation of SLC41A1 variants with changed transport and regulatory characteristics and/or mode of action.
  • Each of these sequences is combined in a mutation library, which comprises at least all sequences of one of the mutation clusters i) to iv). The most preferred embodiment is a mutation library, which combines all four mutation clusters i) to iv).
  • The expected outcome of mutation clusters i, ii and iv is also a definitive confirmation of the SLC41A1 orientation in cytoplasmatic membrane of the cell. As shown by Kolisek et al. (2008), a predicted model which comprises 10 TMs and both N- and C- terminus oriented intracellularly is most likely.
  • Respective mutated SLC41A1 inserts (from cluster i,ii,iii and/or iv) are cloned into cloning vector pUC19, the selected clones are controlled for the proper insert orientation and sequenced for verification purposes. The constructed library is subcloned into respective mammalian expression vector(s) and used for functional screening in an inducible conditional SLC41A1 knockout cell line or into cells of a transgenic conditional knockout animal, which both will be described in more detail below.
  • Also disclosed herein is the use of a protein consisting of an amino acid sequence SEQ ID NO 1 or a protein fragment thereof, in which one or more amino acids are substituted, deleted or added and which has an identity on the amino acid level with SEQ ID NO 1 of at least 90%, preferably at least 95%, more preferably 98%, for the generation and identification of specific antibodies against the cellular Na+/Mg2+ exchanger.
  • Below, the term "identity of at least 90%" always means that this comprises preferably an identity of at least 95%, more preferably an identity of 98%, regardless whether proteins or DNAs are concerned.
  • The related method of identifying antibodies against the cellular Na+/Mg2+ exchanger comprises the steps of:
    • contacting a protein having SEQ ID NO 1 or a protein fragment thereof, in which one or more amino acids are substituted, deleted or added and which has an identity on the amino acid level with SEQ ID NO 1 of at least 90% with a primary antibody under conditions that allow complex formation of the primary antibody with the protein or the protein fragment and
    • detecting the complex.
  • The complex is preferably detected by a secondary antibody. In a preferred embodiment, the primary und/or the secondary antibody comprise(s) a label, which is selected from the group of radioactive label, fluorescent label, enzymatic label, or biotin label.
  • Also disclosed herein is a kit for identifying antibodies against the Na+/Mg2+ exchanger, wherein the kit comprises a protein consisting of an amino acid sequence of SEQ ID NO 1 or a protein fragment thereof, in which one or more amino acids are substituted, deleted or added and which has an identity on the amino acid level with SEQ ID NO 1 of at least 90%.
  • Also disclosed herein is the use of a protein consisting of an amino acid sequence of SEQ ID NO 1 or a protein fragment thereof, in which one or more amino acids are substituted, deleted or added and which has an identity on the amino acid level with SEQ ID NO 1 of at least 90% in a method which uses the protein in order to generate and produce antibodies, preferably monoclonal antibodies, against the Na+/Mg2+ exchanger.
  • Several antibodies are disclosed which selectively bind to the Na+/Mg2+ echanger. Specifically seven antibodies, wherein each specifically recognises one of the following epitopes
    • ESRANAKGVREEDALLENGS,
    • ESDDVSTDRGPAPPSPLKE,
    • LKGNLEMTLASRLSTAAN,
    • STAANIGHMDTPKEL,
    • SSVGGLILDKTVSDPN,
    • PGENSEQAPRRCPSPCTT or
    • ADWMVHWMWGRGLDPDN of SEQ ID NO 1.
  • These antibodies can be used for the detection of the presence, absence and or amount and for the interaction with the cellular Na+/Mg2+ exchanger, which has a protein having the amino acid sequence of SEQ ID NO 1 or a protein fragment thereof, in which one or more amino acids are substituted, deleted or added and which has an identity on the amino acid level with SEQ ID NO 1 of at least 90%. The antibodies may be polyclonal or monoclonal but monoclonal antibodies are preferred.
  • Also disclosed herein is the use of a protein consisting of an amino acid sequence of SEQ ID NO 1 or a protein fragment thereof, in which one or more amino acids are substituted, deleted or added and which has an identity on the amino acid level with SEQ ID NO 1 of at least 90% for the identification of specific protein binding partners that can form stable or transient protein complexes with the cellular Na+/Mg2+ exchanger.
  • The related method of identifying specific protein complex partners for the Na+/Mg2+ exchanger, comprises the steps of:
    • contacting a cell, which contains a protein of amino acid sequence SEQ ID NO 1 or a fragment thereof, in which one or more amino acids are substituted, deleted or added and which has an identity on the amino acid level with SEQ ID NO 1 of at least 90%
      with a candidate substance under conditions that allow for an interaction of the candidate substance with the protein or the fragment,
    • determining the the free or total intracellular Mg2+ concentration or the extracellular Mg2+ concentration, and
    • comparing the Mg2+ concentration as determined with the free or total intracellular Mg2+ concentration or the extracellular Mg2+ concentration in the absence of the candidate substance.
  • Also disclosed herein is a kit for identifying specific protein binding partners forming stable or transient protein complexes with the Na+/Mg2+ exchanger, wherein the kit comprises a protein having an amino acid sequence of SEQ ID NO 1 or a protein fragment thereof, in which one or more amino acids are substituted, deleted or added and which has an identity on the amino acid level with SEQ ID NO 1 of at least 90%.
  • Especially the substances listed below prove suitable as protein complex partner for the Na+/Mg2+ exchanger, which is identical to a protein having an amino acid sequence of SEQ ID NO 1 or a protein fragment thereof, in which one or more amino acids are substituted, deleted or added and which has an identity on the amino acid level with SEQ ID NO 1 of at least 90%:
    • 3-beta-hydroxysteroid-delta(8),delta(7)-isomerase (OS=homo sapiens, UniProt ID Q15125); 3-beta-hydroxysteroid-delta(8),Delta(7)-isomerase OS=Rattus norvegicus, UniProt ID Q9JJ46); ATP synthase subunit a (OS=Cricetulus griseus, UniProt ID P14413); ATP synthase subunit a (OS=homo sapiens, UniProt ID P00846); ATP synthase subunit a (OS=Pan troglodytes, UniProt ID Q9T9W0); ATP synthase subunit a (OS=Tachyglossus aculeatus aculeatus, UniProt ID Q8W9G8); B-cell receptor-associated protein 31 (OS=Homo sapiens, UniProt ID P51572); Immediate early response 3-Interaeting protein 1 (OS=Homo sapiens, UniProt ID Q9Y5U9); Keratinocyte-associated protein 2 (OS=Homo sapiens, UniProt ID Q8N6L1); Kunitz-type protease inhibitor 2 (OS=Homo sapiens, UniProt ID 043291); Peptidyl-prolyl cis-trans isomerase B (OS=Homo sapiens, UniProt ID P23284); Probable ergosterol biosynthetic protein 28 (OS=Pongo abelii, UniProt ID Q5R589); Protein disulfide-isomerase A6 (OS=Homo sapiens, UniProt ID Q15084); Protein YIPF6 (OS=Homo sapiens, UniProt ID Q96EC8); Putative uncharacterized protein CXorf62 (OS=Homo sapiens, UniProt ID Q8N2A0); Signal peptidase complex subunit 1 (OS=Pongo abelii, UniProt ID Q5RF96); Stress-associated endoplasmic reticulum protein 1 (OS=Rattus norvegicus, UniProt ID Q9R2C1); Translocon-associated protein subunit gamma (OS=Homo sapiens, UniProt ID Q9UNL2); Transmembrane protein 147 (OS=Homo sapiens, UniProt ID Q9BVK8); Transmembrane protein 14A (OS=Homo sapiens, UniProt ID Q9Y6G1); Uncharacterized protein C10orf35 (OS=Homo sapiens, UniProt ID Q96D05); Uncharacterized protein C4orf3 (OS=Homo sapiens, UniProt ID Q8WVX3); UPF0414 transmembrane protein C20orf30 (OS=Homo sapiens, UniProt ID Q96A57); UPF0480 protein C15orf24 homolog (OS=Macaca fascicularis, UniProt ID Q4R5V2); and UPF0480 protein C15orf24 (OS=Homo sapiens, UniProt ID Q9NPA0).
  • Preferred molecules out of these are 3-beta-hydroxysteroid-delta(8),delta(7)-isomerase (OS=Homo sapiens, UniProt ID Q15125); B-cell receptor-associated protein 31 (OS=Homo sapiens, UniProt ID P51572); Peptidyl-prolyl cis-trans isomerase (OS=Homo sapiens, UniProt ID P23284) and UPF0480 protein C15orf24 (OS=Macaca fascicularis, UniProt ID Q4R5V2).
  • These SLC41A1 complex-forming partners were screened out by the split-ubiquitin method, which is well known in the art and are listed in detail in Table 1. The ubiquitin-based split protein sensor (USPS) is the method that makes it possible to detect and monitor protein-protein interactions as a function of time, at the natural sites of these interactions in a living cell. Details on the method can be found in Johnsson and Varshavsky, 1994.
  • The more often the screened molecules occur in the list given in Table 1, the higher is their ability of being a binding partner for SLC41A1, i.e. the more clones revealed interaction with SLC41A1. In order to shorten the table, multiple entries were deleted and only the frequency of occurrence (Occurrence) is indicated as a number. The singletons listed in Table 2 are the weakest partners. They are organized from the strongest to the weakest. Table 1
    protein name (description) uniprot ID occurrence
    3-beta-hydroxysteroid-Delta(8),Delta(7)-isomerase OS=Homo sapiens GN=EBP PE=1 SV=3 Q15125 24
    3-beta-hydroxysteroid-Delta(8),Delta(7)-isomerase OS=Rattus norvegicus GN=Ebp PE=2 SV=3 Q9JJ46 1
    ATP synthase subunit a OS=Cricetulus griseus GN=MT-ATP6 PE=3 SV=1 P14413 1
    ATP synthase subunit a OS=Homo sapiens GN=MT-ATP6 PE=1 SV=1 P00846 1
    ATP synthase subunit a OS=Pan troglodytes GN=MT-ATP6 PE=3 SV=1 Q9T9W0 1
    ATP synthase subunit a OS=Tachyglossus aculeatus aculeatus GN=MT-ATP6 PE=3 SV=1 Q8W9G8 1
    B-cell receptor-associated protein 31 OS=Homo sapiens GN=BCAP31 PE=1 SV=3 P51572 10
    Immediate early response 3-interacting protein 1 OS=Homo sapiens GN=IER3IP1 PE=1 SV=1 Q9Y5U9 3
    Keratinocyte-associated protein 2 OS=Homo sapiens GN=KRTCAP2 PE=1 SV=1 Q8N6L1 2
    Kunitz-type protease inhibitor 2 OS=Homo sapiens GN=SPINT2 PE=1 SV=2 043291 2
    Peptidyl-prolyl cis-trans isomerase B OS=Homo sapiens GN=PPIB PE=1 SV=2 P23284 5
    Probable ergosterol biosynthetic protein 28 OS=Pongo abelii PE=2 SV=1 Q5R589 2
    Protein disulfide-isomerase A6 OS=Homo sapiens GN=PDIA6 PE=1 SV=1 Q15084 11
    Protein YIPF6 OS=Homo sapiens GN=YIPF6 PE=2 SV=2 Q96EC8 2
    Putative uncharacterized protein Cxorf62 OS=Homo sapiens GN=Cxorf62 PE=2 SV=1 Q8N2A0 2
    Signal peptidase complex subunit 1 OS=Pongo abelii GN=SPCS1 PE=3 SV=1 Q5RF96 2
    Stress-associated endoplasmic reticulum protein 1 OS=Rattus norvegicus GN=Serp1 PE=1 SV=2 Q9R2C1 4
    Translocon-associated protein subunit gamma OS=Homo sapiens GN=SSR3 PE=1 SV=1 Q9UNL2 4
    Transmembrane protein 147 OS=Homo sapiens GN=TMEM147 PE=1 SV=1 Q9BVK8 2
    Transmembrane protein 14A OS=Homo sapiens GN=TMEM14A PE=2 SV=1 Q9Y6G1 2
    Uncharacterized protein C10orf35 OS=Homo sapiens GN=C10orf35 PE=1 SV=1 Q96D05 2
    Uncharacterized protein C4orf3 OS=Homo sapiens GN=C4orf3 PE=2 SV=1 Q8WVX3 2
    UPF0414 transmembrane protein C20orf30 OS=Homo sapiens GN=C20orf30 PE=1 SV=1 Q96A57 3
    UPF0480 protein C15orf24 homolog OS=Macaca fascicularis GN=QtsA-20627 PE=2 SV=1 Q4R5V2 1
    UPF0480 protein C15orf24 OS=Homo sapiens GN=C15orf24 PE=1 SV=1 Q9NPA0 4
  • Further Singletons are listed below in table 2. Table 2
    Protein name (description) UniProt ID
    ADP-ribosylation factor-like protein 6-interacting protein 4 OS=Homo sapiens GN=ARL6IP4 PE=1 SV=1 Q66PJ3
    Elongation of very long chain fatty acids protein 4 OS=Homo sapiens GN=ELOVL4 PE=2 SV=1 Q9GZR5
    Proteolipid protein 2 OS=Homo sapiens GN=PLP2 PE=1 SV=1 Q04941
    Putative P2Y purinoceptor 10 OS=Homo sapiens GN=P2RY10 PE=2 SV=1 000398
    Transmembrane protein 106C OS=Homo sapiens GN=TMEM106C PE=2 SV=1 Q9BVX2
    Solute carrier family 35 member B1 OS=Homo sapiens GN=SLC35B1 PE=2 SV=1 P78383
    Thioredoxin-related transmembrane protein 1 OS=Homo sapiens GN=TMX1 PE=1 SV=1 Q9H3N1
    Transmembrane emp24 domain-containinq protein 10 OS=Homo sapiens GN=TMED10 PE=1 SV=2 P49755
    Putative uncharacterized protein C3orf66 OS=Homo sapiens GN=C3orf66 PE=5 SV=1 Q6ZWE1
    Probable phospholipid-transporting ATPase ID OS=Homo sapiens GN=ATP8B2 PE=1 SV=2 P98198
    Signal recognition particle 9 kDa protein OS=Homo sapiens GN=SRP9 PE=1 SV=2 P49458
    Zinc transporter ZIP13 OS=Rattus norvegicus GN=SIc39a13 PE=2 SV=1 Q2M1K6
    Phosphatidate phosphatase PPAPDC1B OS=Mus musculus GN=Ppapdc1b PE=2 SV=1 Q3UMZ3
    WW domain-binding protein 11 OS=Rattus norvegicus GN=Wbp11 PE=2 SV=1 Q5PQQ2
    Emerin OS=Homo sapiens GN=EMD PE=1 SV=1 P50402
    Transmembrane 6 superfamily member 1 OS=Homo sapiens GN=TM06F1 PE=2 SV=2 Q9BZW5
    C-4 methylsterol oxidase OS=Pongo abelii GN=SC4MOL PE=2 SV=1 Q5R574
    Single-stranded DNA-binding protein 2 OS=Homo sapiens GN=SSBP2 PE=1 SV=2 P81877
    Putative Uncharacterized protein C14orf165 OS=Homo sapiens GN=C14orf165 PE=4 SV=1 Q86U02
    Zinc finger protein 714 OS=Homo sapiens GN=ZNF714 PE=2 SV=1 Q96N38
    Calcium release-activated calcium channel protein 1 OS=Homo sapiens GN=ORAI1 PE=1 SV=2 Q96D31
    Transmembrane BAX inhibitor motif-containing protein 4 OS=Homo sapiens GN=TMBIM4 PE=1 SV=2 Q9HC24
    NADH-ubiquinone oxidoreductase chain 3 OS=Homo sapiens GN=MT-ND3 PE=1 SV=1 P03897
    ADP-ribosylation factor-like protein 6-interacting protein 6 OS=Homo sapiens GN=ARL6IP6 PE=1 SV=1 Q8N065
    Ribonuclease kappa OS=Mus musculus GN=Rnasek PE=2 SV=1 Q8K3C0
    Wiskott-Aldrich syndrome protein homolog OS=Mus musculus GN=Was PE=1 SV=1 P70315
    Cytochrome c oxidase subunit 2 OS=Homo sapiens GN=MT-CO2 PE=1 SV=1 P00403
    Melanoma-associated antigen D1 OS=Mus musculus GN=Maged1 PE=1 SV=1 Q9QYH6
    Probable low affinity copper uptake protein 2 OS=Homo sapiens GN=SLC31A2 PE=2 SV=1 015432
    Protein cornichon homolog 4 OS=Pongo abelii GN=CNIH4 PE=2 SV=1 Q5R9M4
    Mitochondrial carrier homolog 2 OS=Pongo abelii GN=MTCH2 PE=2 SV=2 Q5R5M0
    Squalene synthetase OS=Pongo abelii GN=FDFT1 PE=2 SV=1 Q5R6U3
    RING finger protein 114 OS=Pan troglodytes GN=RNF114 PE=2 SV=1 Q6J212
    Interferon alpha-inducible protein 6 OS=Homo sapiens GN=IFI6 PE=2 SV=2 P09912
    Protein tyrosine phosphatase receptor type C-associated protein OS=Homo sapiens GN=PTPRCAP PE=1 SV=1 Q14761
    Protein FAM192A OS=Homo sapiens GN=FAM192A PE=1 SV=1 Q9GZU8
    Intercellular adhesion molecule 2 OS=Homo sapiens GN=ICAM2 PE=1 SV=2 P13598
    Leptin receptor gene-related protein OS=Homo sapiens GN=LEPROT PE=1 SV=1 O15243
  • Also disclosed herein is the use of said molecules as protein complex partners for the Na+/Mg2+ exchanger thereby regulating or modifying its function. Additionally, these specific protein binding partners are intended for the use in therapeutical applications, especially for the treatment of the diseases mentioned below (Table 4).
  • In the above tables 1 and 2 underlined names indicate highly connected proteins, found in the majority of DUALmembrane SU-YTH screens. Highly connected proteins include chaperones, stress-induced endoplasmatic reticulum proteins, components of the translocon machinery and proteins involved in membrane protein sorting and transport.
  • The most prominent candidate binding partner is 3-beta-hydroxysteroid-Delta(8),Delta(7)-isomerase, an enzyme which catalyzes the conversion of Delta(8)-sterols to their corresponding Delta(7)-isomers. In the screening 24 clones were identified where 3-beta-hydroxysteroid-Delta(8),Delta(7)-isomerase interacted with SLC41A1. Defects in 3-beta-hydroxysteroid-Delta(8),Delta(7)-isomerase are the cause of chondrodysplasia punctata also known as Conradi-Hunermann-Happle syndrome (Derry et al, 1999). As for the other candidate binding partners identified, the nature of interaction between SLC41A1 and 3-beta-hydroxysteroid-Delta(8),Delta(7)-isomerase is unknown. Its identification will help to understand the link between disturbed cholesterol metabolism and magnesium insufficiency in mammals.
  • Ten clones revealed interaction between SLC41 and B-cell receptor associated-protein 31. It is expected to play a role in anterograde transport of membrane proteins from the endoplasmic reticulum to the Golgi. It may be also involved in CASP8-mediated apoptosis.
  • The third strongest SLC41A1-binding candidates were Peptidyl-prolyl cis-trans isomerase and UPF0480 protein C15orf24.
  • The rest of the identified SLC41A1 binding partners, found in more than only one clone, is merely represented by poorly characterized or uncharacterized transmembrane proteins, proteins involved in mediating the cellular response to mitogenic signal (immediate early-response 3-interacting protein; SABiosciences) and endoplasmatic reticulum-located probable ergosterol biosynthetic protein 28, transmembrane protein 147 and APH-1-similar core component of nicalin-nomo complexes. One interesting candidate is the Kunitz type protease inhibitor 2. When unfunctional, the protein cause a rare, inherited diarrhea of infancy named congenital secretory sodium diarrhea type 3 (DIAR3) or congenital sodium diarrhea (CSD).
  • Interesting binding partners among the singletons are: probable low affinity copper uptake protein 2, mitochondrial carrier homologue 2 (transporter of an unknown solute), calcium release-activated calcium channel protein 1, zinc transporter ZIP13 and solute carrier family 35 member B1 which is likely to be an endoplasmatic reticulum located sugar transporter (Uniprot).
  • Further substances, which either increase or decrease the SLC41A1-related Na+/Mg2+ exchanger activity and are thus called modulators, are given in Table 3. These substances modulate SLC41A1-related Na+/Mg2+ exchanger activity either by direct interaction with structural components of the SLC41A1 molecule or by the induction of cellular signal cascades resulting in modifications of the SLC41A1 protein. Therefore, some of these modulators are hormones or elements of cellular signal cascades. Main sites for posttranslational modifications and thus, regulation/modulation of SLC41A1-related Na+/Mg2+ exchanger activity were the following:
    • Glycosylation side: amino acid residue N-475
    • cAMP-dependent PK phosphorylation sides: S-157, S-308, S-393, T-508
    • PK C phosphorylation sides: T-80, T-167, T-387, S-407
    • Casein kinase II motif at residue S-317
    • Myristoylation sequence at G-143
    Table 3
    Modulator Chemical name Molecular weight (g/mol) Dosage (µM) Effect on Na+/Mg2+ exchanger Further Effect on
    Imipramine 10,11-Dihydro-N,N-dimethyl-5H-dibenz-[b,f]azepin-5-propanamin 280.4 250 Inhibition Na+ channel blocker
    Quinidin (2-ethenyl- 4-azabicyclo [2.2.2] oct- 5-yl)- (6-methoxyquinolin- 4-yl)-methanol 324.4 100 Inhibition K+ channel blocker
    DIDS 4',4'-diisothiocyanatodihydrostil bene-',2'-disulfonic acid, disodium salt 500.48 100 Inhibition Cl- channel inhibitor, Anion exchanger inhibitor
    Lidocaine 2-(diethylamino)-N-(2,6-dimethylphenyl)-acetamide 234.34 Inhibition Na+ channel blocker
    KB-R7943 2-(2-(4-(4-nitrobenzyloxy)-phenyl)ethyl)isothiourea methanesulfonate 427.50 Inhibition Na+/Ca2+ exchanger inhibitor
    SEA0400 2-[4-[(2,5-difluoro-phenyl)methoxy]phenoxy]-5-ethoxyaniline 371.38 Inhibition Na+/Ca2+ exchanger inhibitor
    Benzamil 3,5-diamino-N-[(1E)-amino(benzylamino)methyl idene]- 6-chloropyrazine-2-carboxamide 319.75 10 µM Inhibition Na+/Ca2+ exchanger inhibitor
    Amiloride 3,5-diamino-6-chloro-N-(diaminomethylidene)pyraz ine-2-carbox-amide 229.63 5 - 100µM Inhibition Na+/H+ exchanger (NHE) inhibitor
    Nifedipine 3,5-dimethyl 2,6-dimethyl-4-(2-nitrophenyl)-1,4-di-hydropyridine-3,5-di-carboxylate 346.36 Inhibition Ca2+ channel blocker
    Angiotensin II (via AT 1 receptor) 100 nM activation Vasoactive agent, vasoconstriction
    Aldosterone activation
    Glucocorticoids Dexamethason 392.46
    Catecholamines, β-adrenergic agonists α1-adrenergic agonists Norepinephrine, isoproterenol activation
    cAMP and cAMP analogues 8-Cl-cAMP; dibutyryl-cAMP 100 µM activation
    Rp-cAMP inhibition
    Forskolin [(3R,4aR,5S,6S,6aS, 10S,1 0aR,10bS)-3-ethenyl-6,10,10b-trihydroxy-3,4a,7,7,10a-pentamethyl-1-oxo-5,6,6a,8,9,10-hexahydro-2H-benzo[f]chromen-5-yl] acetate 410.50 activation
    Cardiotonic steroids Ouabain, rostafuroxin, bufalin, oleandrin, inhibition
    (glycosides) marinobufagenin, UNBS-1450
    Glucagon activation
    Insulin (via insulin receptor) 200 µU/ml inhibition
    400 µU/ml
    Genistein 5,7-dihydroxy-3-(4-hydroxyphenyl)chromen-4-one 270.23 10 µM inhibition Tyrosine kinase inhibitor
    Mitogens Bombesin, epidermal growth factor (EGF) inhibition
    Taurine 2-aminoethanesulfonic acid 10-20 mM inhibition
    Organic polycations
    Spermine, spermidine N', N'-bis(3-aminopropyl)-butane-1,4-diamine, N-(3-aminopropyl)butane-1,4-diamine
    PhTX-343 S-N-[3-[[4-[(3-Aminopropyl)amino]butyl]-amino]propyl]-4-hydroxy-alpha-[(1-oxobutyl)amino]-benzenepropanamide tris-trifluoroacetate (Organic polycation) Derivative of the naturally occuring polyamine toxin philantotoxin
    Agmatine 2-(4-aminobutyl)guanidine 130.19 Polyamine in neurons
    L-NMMA NG-monomethyl-L-arginine 100 µM inhibition NO inhibitor
    ODQ 1H-[1,2,4]oxadiazolo[4,3-α]quinoxaline-1 10 µM inhibition inhibitor of NO-sensitive guanylate cyclase
    NOR1 (E)-4-methyl-2-[(E)-hydoxyimino]-5-nitro-6-methoxy-3-hexenamide 100 µM activation NO donor
    Calphostin C 1-[3,10-dihydroxy-12-[2-(4-hydroxyphenoxy)-carbonyloxypropyl]-2,6,7,11-tetramethoxy-4,9-dioxoperylen-1-yl]propan-2-yl benzoate 790.76 1 µM inhibition PK C inhibitor
    Phorbol 12,13-dibutyrate 504.61 1µM activation PK C activator
    cGMP and cGMP analogues 8-bromo-cyclic GMP 100 µM activation
    PGE2 (Z)-7-[(1R,2R,3R)-3-hydroxy-2-[(E,3S)-3-hydroxyoct-1-enyl]-5-oxocyclopentyl]hept-5-enoic acid 352.47 100 nM activation
    Indomethacin 2-[1-(4-chlorobenzoyl)-5-methoxy-2-methylindol-3-yl]acetic acid 357.79 10µM activation Arachidonic acid metabolism inhibitor
    Arachidonic acid (5Z,8Z,11Z,14Z)-icosa-5,8,11,14-tetraenoic acid 304.47 30 µM activation
  • Particularly imipramine, quinidin, cyclic adenosine monophosphate (cAMP) analogues, β-adrenergic and α1-adrenergic agonists and the hormones angiotensin II and insulin showed strong modulating effects. It could be demonstrated that replacement of extracellular Na+ with N-methyl-D-glutamine (NMDG) or application of the unspecific Na+/Mg2+ exchanger inhibitors imipramine and quinidin strongly decrease SLC41A1-mediated efflux. Also, it was found that SLC41A1-mediated Mg2+ efflux is stimulated by db-cyclic adenosine monophosphate (db-cAMP), adrenergic receptor agonists and the hormone angiotensin II. In contrast, an inhibitory effect was observed after application of insulin.
  • In view of the importance of the Na+/Mg2+ exchanger activity to cell metabolism, viability and proliferation, we believe that SLC41A1 represent an ideal target for designing and /or identifying molecules that block or stimulate these processes.
    Therefore, the above mentioned modulators (Table 3), especially imipramine, quinidin, DIDS, lidocaine, nifedipine, benzamil, KB-R7943, SEA0400, amiloride, agonists or antagonists of receptors for glucocorticoids, aldosterone, angiotensin II, insulin, glucagon, PGE2 and catecholamines (β-adrenergic and/or α1-adrenergic agents), cAMP or cGMP analogues, taurine, genistein, calphostin C, phorbol 12,13-dibutyrate, L-NMMA, NOR1, ODQ, forskolin, indomethacin, arachidonic acid, spermine, spermidine, PhTX-343, agmatine, cardiotonic steroids (e.g. ouabain, rostafuroxin, bufalin, oleandrin, marinobufagenin, UNBS-1450) and of mitogens (e.g. bombesin, EGF),more preferably imipramine, quinidin, cAMP and its analogues, adrenergic agonists and molecules that mimic the effects of angiotensin II and of insulin, can be used to modulate the function, metabolism, proliferation and growth of Na+/Mg2+ exchanger expressing cells. The above named modulators are also intended for the use as therapeutic agents.
  • The above-mentioned antibodies, protein binding partners and the above named modulators or their analogues or agents acting as agonists or antagonists of these modulators are believed to be useful for treating patients having conditions that are characterizid by increased or decreased Mg2+ efflux via their Na+/Mg2+ exchanger expressing cells. Such conditions of disturbed or dysregulated Na+/Mg2+ exchanger activity have been described in various disease complexes such as cardiovascular disorders, metabolic and endocrine dysfunctions, neurologic diseases, cancer, pre-eclampsia, eclampsia, fetal growth retardation and premature labor.
  • Special diseases out of these disease complexes, which the present invention is focused on, are listed in Table 4. Diseases, on which the disclosure is preferably focused on, are hypertension, ischemic heart disease, myocardial infarction, cardio-vascular dysfunctions in relation to diabetes mellitus, pre-eclampsia, eclampsia, diabetes mellitus (type II), non-insulin-dependent diabetes, cystic fibrosis, morbus parkinson, stroke, prostate cancer as well as aggressive behaviour or high stress sensitivity. These diseases are marked in Table 4 (bold). Table 4
    Disease complex Disease Role of Na+/Mg2+ exchanger Pathogenic factors Relevancy/strength of evidence
    Cardiovascular Hypertension (high blood pressure) hyperactivity Potentiation of platelet aggregation, Loss of Mg2+ from heart muscle cells, hepatocytes, platelets and smooth vascular muscle cells, Increased sensitivity to vasoconstrictors ***** (5)
    ischemic heart disease, myocardial infarction
    congestive heart failure
    atherosclerosis
    cardiac arrhythmias
    sudden cardiac death
    Cardiovascular dysfunction in relation to diabetes mellitus
    Pre-eclampsia, Eclampsia (strongly related to pregnancy)
    Metabolism Endocrine dysfunction Diabetes mellitus (type I) Non-insulin-dependent diabetes (type II) hyperactivity **** (4)
    Hyoeraldosteronism
    Hyperthyroidism/ hypothyroidism I ncrease/loss of Mg2+ in/from heart muscle cells
    Mukoviszidose Cystic fibrosis hyperactivity
    Neurology/ behaviour Parkinson disease, Morbus Parkinson ** (2)
    Aggressive behaviour, high stress sensitivity hyperactivity
    stroke
    Fetal growth retardation, premature labor ** (2)
    cancer Prostate cancer Upregulated ** (2)
    hyperactivity
  • In Table 4, the diseases are listed from top to bottom according to the present evidence that their pathogenesis is related to an increased or decreased Mg2+ efflux via Na+/Mg2+ exchanger expressing cells. The strength of evidence is indicated by stars in the last column. Cardiovascular diseases have the strongest evidence that they are related to the Na+/Mg2+ exchanger. Thus, the treatment of cardiovascular diseases is the most relevant application for the present invention, i.e. for the antibodies, the special protein binding partners as outlined above or for the specific modulators outlined above.
  • The nucleotide sequence of SEQ ID NO 3 (cDNA) is of special use for the gene therapy of these diseases, i.e. for the treatment of a disease from the group of cardiovascular diseases, metabolic or endocrine dysfunctions, neurological diseases, cancer, pre-eclampsia, eclampsia, fetal growth retardation and premature labor, preferably for the treatment of hypertension, ischemic heart disease, myocardial infarction, cardiac dysfunction in relation to diabetes mellitus, pre-eclampsia, eclampsia, diabetes mellitus (type I), non-insulin-dependent diabetes, mukoviszidose, cystic fibrosis (type II), parkinson disease, morbus parkinson, stroke, prostate cancer as well as aggressive behaviour or high stress sensitivity. The same applies to a fragment of SEQ ID NO 3, in which one or more nucleotides are replaced, deleted or added and which still has an identity on the nucleotide level of at least 90%.
  • Also disclosed herein is a model based on a non-human mammal, preferably a mouse, having an inducible conditional system leading to a functional knock out of SLC41A1, i.e. a genetically modified conditional knock out non-human mammal, which is characterized in that the SCL41A1 gene is inducible conditionally knockable (knock out), i.e. it can be knocked-out in a time (e.g., at embryonic, young or adult stage of development) or tissue-specific (particular organ or cell type) manner. In the following, the mouse is referred to because this is the preferred non-human mammal.
  • The mouse model was developed on the basis of the gene sequence of SLC41A1 (SEQ ID NO 3, GenBank Accession No. NM_173854).
  • A SLC41A1 inducible conditional KO mouse was developed by homologous recombination using 129 embryonic stem cells. This mouse line was then crossed with Cre recombinase (CR) or ligand-dependent Cre recombinase (LCR) expressing mice for the generation of tissue specific or time dependent targeted inactivation of murine SLC41A1.
  • LCR are fusion proteins between Cre and the ligand-binding domains (LBDs) of steroid receptors. The LBD has been mutated so that it does not respond to its natural ligand yet binds a synthetic ligand. As an example may serve tamoxifen-activated CreERT recombinase (modified pacman), a fusion of Cre with a mutated estrogen receptor (ER) LBD, that responds specifically to the synthetic drug 4-hydroxytamoxifen (OHT) but not to β-estradiol. In the absence of OHT, the recombinase is located in the cytoplasm. Binding of OHT to the LBD results in the translocation of the recombinase into the nucleus where it can recombine its IoxP substrates (Feil R., 2007). The LCR system is widely used for the generation of inducible conditional KO mice in experiments with time dependent settings but also in those with demand for tissue specificity. Mouse lines expressing CreERT2 recombinase in tissue specific manner are listed below and the newly developed conditional KO SLC41A1 mouse can be bread with one of the available CreERT2 mouse lines in dependence of specific experimental needs.
  • Examples of mouse lines expressing the CreERT2 recombinase in soecific tissues:
    • Bone (osteoblasts Col1a1-CreERT2 tg Collagen 1 α1 chain and odontoblasts)
    • Endothelium (Tie2-CreERT2 tg Tie2 receptor tyrosine kinase)
    • Epithelium:
    • Intestinal epithelium Vil-CreERT2 tg Villin el
    • Internal epithelial organs K18-CreERT2 tg Keratin 18
    • Renal epithelium KspCad-CreERT2 tg Kidney-specific cadherin Lantinga-van
    • Fat tissue (adipocytes) aP2-CreERT2 tg Adipocyte fatty acid binding protein
    • Liver (hepatocytes) SA-CreERT2 ki Serum albumin
    • Nervous system:
    • Astrocytes GFAP-CreERT2 tg Glial fibrillary acidic protein
    • GLAST-CreERT2 ki Astrocyte-specific glutamate transporter
    • Neural stem cells Nes-CreERT2 tg Nestin
    • Schwann cells and PLP-CreERT2 tg Proteolipid protein
    • oligodenrocytes
    • Schwann cells P0Cx-CreERT2 tg P0 fused to connexin 32
    • Skin:
    • Keratinocytes K5-CreERT2 tg Keratin 5
    • K14-CreERT2 tg Keratin 14
    • Melanocytes Tyr-CreERT2 tg Tyrosinase
    • Tyr-CreERT2 tg Tyrosinase
    • Widespread Rosa26-CreERT2 ki Rosa26.
    • Muscle:
    • Skeletal muscle HAS-CreERT2 tg (PAC) Skeletal muscle α-actin
    • Smooth muscle SM-CreERT2 ki SM22α
  • The transgenic conditional knock out mouse is characterized in that the genome comprises a construct, which allows conditional knock out of the SCL41A1. The term "construct" refers to a polynucleotide, which was exogenously inserted into the genom of the animal. The animal can be heterozygous or homozygous but is preferably homozygous with respect to the knock out gene (-/-). The construct comprises the SCL41A1 gene of SEQ ID NO 4 (murine gene) flanked by two IoxP DNA sequence elements (IoxP sites), i.e. they are preferably situated 3' and 5' of the SCL41A1 gene.
  • Isolated cells from various tissues allow further investigation of the function of SCL41A1. Thus, also disclosed herein is a transgenic inducibleconditional knock out cell or cell line. Cells can be taken from the transgenic inducible conditional knock out non-human mammal or can originate from separate cell lines.
  • If cells from the transgenic animal are concerned, it is preferred to use primary cell cultures (primocultures) from the heart (cardiomyocytes), from the vasculature (e.g. aorta), from the liver (hepatocytes) or from the brain or CNS/microglial cells of these animals.
  • If cells or cell lines of other origin are concerned, it is preferred to use stable human cell lines, more preferably stable human embryonic kidney cell line (ACC-305/DSMZ) with inducible conditionally knocked out SLC41A1 gene, HEK293 (hSLC41A1-/-) or stable human B-cell precursor leukemia cell line (ACC-128/DSMZ) with inducible conditionally knocked out SLC41A1 gene, NALM-6 (hSLC41A1-/-). Cell lines will be generated according to the technique described by Shibahara Kei-ichi et al..
    To generate inducible conditionally knocked out SLC41A1 cells or cell lines, Mammalian vectors, which comprise mutated forms of human SLC41A1 are transferred or electroporated into these primocultured cells of or into cells of the stable human cell lines, resulting in cells having in their genome mammalian vectors with a mutated human SLC41A1. The mutated forms of human SLC41A1 are as described above with respect to the mutation libraries, i.e. each of these mutated sequences can be present in the vector.
  • Description of the Figures
  • Figure 1
    shows Mg2+ Efflux in dependence of the Mg2+ concentration of the loading medium.
    1. A: Representative original recordings of [Mg2+]i changes after re-suspension of -tet and +tet HEK293-(SLC41A1) cells in Mg2+-free, Na+-containing medium. Before measurements cells had been loaded with Mg2+ by incubating them in a high (10mM)-Mg2+ solution for 20 min.
    2. B: Magnitude of mean [Mg2+]i changes for -tet and +tet HEK293-(SLC41A1) cells pre-loaded in solutions containing 0, 2, 5 or 10 mM Mg2+. N = 10 single experiments for each Mg2+ concentration; * P<0.01; ** P<0.001.
    Fig. 2
    shows the effect of complete substitution of extracellular Na+ by NMDG on SLC41A1-dependent Mg2+ efflux. A: Representative original recordings of [Mg2+]i changes after re-suspension of -tet and +tet HEK293-(SLC41A1) cells in Mg2+-free, Na+-containing or in Mg2+- and Na+-free medium. Before measurements cells had been loaded with Mg2+ by incubating them in a high (10mM)-Mg2+ solution for 20 min. B: Effect of Na+substitution on [Mg2+]i of -tet and +tet HEK 293-(SLC41A1) cells before and after a 20 min incubation in completely Mg2+-free medium. N = 9 single experiments per condition; ** P<0.001. C: Effect of Na+ substitution on SLC41A1-dependent Mg2+ extrusion measured as decrease of [Mg2+]i. N = 9; P<0.001.Mg2+
    Fig.3:
    shows the effect of imipramine and of quinidin on SLC41A1-dependent Mg2+ efflux. A: Representative original recordings of [Mg2+]i changes after re-suspension of +tet HEK293-(SLC41A1) cells in Mg2+-free, Na+-containing media with or without imipramine (250 µM). Before measurements cells were loaded with Mg2+ by incubating them in a high (10mM)-Mg2+ solution for 20 min. B: Summary of the results obtained from 8 single measurements per condition. ** P<0.001. C: Summary of the effect of quinidin (100 µM) on Mg2+]i changes observed after re-suspension of +tet HEK293-(SLC41A1) cells in Mg2+-free, Na+-containing media. Before measurements cells were loaded with Mg2+ by incubating them in a high (10mM)-Mg2+ solution for 20 min. N = 3 single measurements per condition.
    Figure 4:
    shows the effect of dB-cAMP on [Mg2+]i decrease in +tet HEK293-(SLC41A1) cells The [Mg2+]i decreased observed without and with dB-cAMP (100 µM) is shown. Bars represent means ± SE from 6 (0 mM-loading), to 10 (2 mM and 10 mM-loadingl) single measurements.
    Figure 5:
    Original traces showing the effect of imipramine (250 µM) and of dB-cAMP (100 µM) on the proliferation of HEK293-(SLC41A1) cells. The respective agents are added 24 hours after seeding. Increase or decrease of the cell index reflects a higher or lower proliferation rate, respectively. Red curve: control; blue curve: imipramine; green curve = dB-cAMP.
    Figure 6:
    Effect of insulin on the SLC41A1-dependent Mg2+ efflux in -tet and +tet HEK293-(SLC41A1) cells.. [Mg2+]i changes after re-suspension of -tet and +tet HEK293-(SLC41A1) cells in Mg2+-free, Na+-containing media without or with insulin (200 µU/ml and 400 µU/ml) are shown. Before measurements cells were loaded with Mg2+ by incubating them in a high (10mM)-Mg2+ solution for 20 min. N = 3 single measurements per condition.
  • Below, the invention is outlined in more detail by means of example.
  • Examples Material and Methods: HEK293-(FLAG-SLC41A1) cell line; growth media and culture conditions
  • Preparation (cloning protocol) and culture conditions of tetracycline-(tet)-inducible HEK293-(FLAG-SLC41A1) cell line were previously described by Kolisek et al. [2008].
  • Wesfern Blot Analysis
  • Non-induced (-tet) and induced (+tet, 15-18 h) HEK293-(FLAG-SLC41A1) cells (107 cells ml-1) were lysed for 30 min at 4 °C in Tris buffer (pH 7.5) containing 1% Triton X-100 (Bio-Rad) and protease inhibitors (Perbio-ThermoFisher). Total lysate proteins had been resolved by 10% SDS-PAGE, transferred to polyvinylidene difluoride membranes, and immunodecorated with anti-FLAG antibody coupled to HRP (Invitrogen), or with antibody to -actin (AbCam, Cambridge, UK) conjugated to GAM HRPlinked antibody (Jackson ImmunoResearch Laboratories, West Grove, PA). The same samples were immunoprecipitated by M2 anti-FLAG (Sigma) or isotype control, resolved by 10% SDS-PAGE, and transferred to polyvinylidene difluoride membranes. The membrane was immunoblotted with M2 anti-FLAG (Sigma) and GAM-_-HRP (SBA, Birmingham, AL). Membranes were developed by enhanced chemical luminescence (ECL) (Amersham Biosciences).
  • Determination of free intracellular Mg2+ in +tet and -tet HEK293-(SLC41A1) cells by mag-fura 2 FF-Spectrofluorometry
  • The uninduced (-tet) and induced (+tet) HEK293-(SLC41A1) cells were rinsed twice with ice-cold, completely divalent-free Dulbecco's phosphate-buffered saline (DPBS), detached by use of Hytase (Perbio Science, Bonn, Germany), centrifuged, washed twice in completely Ca2+- and Mg2+-free Hank's balanced solution (CMF-HBS, PAN Biotech, Aidenbach, Germany) complemented with 10 mmol.l-1 HEPES and 1.36 mmol.l-1 L-glutamine (CMF-HBS+) and finally re-suspended in the same solution. Loading of cells with 7.5 µmol.l-1 mag-fura 2 AM (Molecular Probes, Eugene, OR USA) was performed for 30 minutes at 37°C in the presence of loading facilitator Pluronic F-127 (Molecular probes). After being washed in CMF-HBS+, cells were incubated for a further 30 minuts at 37°C to allow for complete deesterification of the fluorescence probe, washed twice in CMF-HBS+ to remove extracellular mag-fura 2 and stored in CMF-HBS supplemented with 10 mmol.l-1 HEPES, 5 mmol.l-1 glucose and 0.4 mmol.l-1 Mg2+ until being Mg2+-loaded before measurements of the Mg2+ efflux capacity. The latter was determined as the intracellular Mg2+ concentration ([Mg2+]i) decrease over a 20-min period. Directly before the efflux experiments, -tet and +tet HEK293-(SLC41A1) were Mg-loaded by a 20-min incubation in CMF-HBS+ containing 0, 2, 5 or 10 mmol.l-1 Mg. An extracellular Mg2+ concentration ([Mg2+]e) of 10 mmol.l-1 was selected for all further experiments. After loading, the remaining extracellular Mg2+ was removed by washing the cells twice in CMF-HBS+ and, after re-suspending the cells in CMF-HBS+, measurements were started. To determine the influence of varying extracellular [Na+], measuring solutions were prepared by diluting the CMF-HBS+ containing 145 mM Na+ with appropriate volumes of a completely Na+- and Mg2+-free PBS. To investigate the effect of HCO3 - on Mg2+ efflux, completely Ca2+- and Mg2+-free medium 199 completed with 10 mmol.l-1 HEPES and 1.36 mmol.l-1 L-glutamine (CMF-M199+) with Hank's (0.35 g/l NaHCO3) or Earl's (2.2 g/l NaHCO3) salts were used during Mg-loading and measurements. To ensure a stable pH value, the high- HCO3 - media were pre-equilibrated with 95% air-5% CO2.
  • Measurements were made at 37°C in 3 ml cuvettes containing 2 ml cell suspension (CMF-HBS+ or CMF-M199+ with a cytocrit of 10 %) under stirring. [Mg2+]i was determined by measuring the fluorescence of the probe-loaded cells in a spectrofluorometer (LS50-B, PerkinElmer Life Science, Wellesley, MA USA) by using the fast filter accessory, which allowed fluorescence to be measured at 20-ms intervals with excitation at 340 and 380 nm, and emission at 515 nm. [Mg2+]i values were calculated from the 340/380-nm ratio according to the formula of Grynkiewicz et al. (1981) by using the software FL WinLab version 4.0 (PerkinElmer Life Science). A dissociation constant of 1.5 mmol.l-1 for the mag-fura 2/Mg2+ complex was used for calculations; minimum (Rmin) and maximum (Rmax) ratios were determined at the end of each experiment by using digitonin. Rmax was found by the addition of 25 mmol.l-1 MgCl2 in the absence of Ca2+, whereas Rmin was obtained by addition of 50 mmol.l-1 EDTA, pH 7.2, to remove all Mg2+ from the solution. For data evaluation, 10-second data sets each were averaged at the beginning of the measurement and then always after 50 seconds. The final [Mg2+]i was determined as the mean [Mg2+]i of the last 10 seconds of the measurement. Thus, for the calculation of any given [Mg2+]i, 500 data points were used. If not otherwise stated, data are presented as means ± standard error (SE). Table 5: List of special media
    Name Remarks Producer
    -Cl-, -Mg2+, -Ca2+ Hank's medium Cl-, Mg2+, Ca2+ free medium (Cl- has been substituted with glutamate)
    Completely Mg2+ - and Na+-free PBS Without Ca2+ /Mg2+, Na+ substituted by N-methyl-D-glucamine (NMDG) No. LZ 1825, Biochrom, Berlin, Germany
    Ca2+- and Mg2+-free medium 199 Hanks Without Ca2+ /Mg2+ and phenol red No. FZ 0635, Biochrom, Berlin, Germany
    Ca2+- and Mg2+ -free medium 199 Earl Without Ca2+ /Mg2+ and phenol red No. FZ 0615, Biochrom, Berlin, Germany
    Table 6: List of inhibitors
    Name Formula MW (g.mol-1) Final concentration (mol.l-1) Producer Inhibitor of:
    Imipramine 10,11-Dihydro-N,N-dimethyl-5H-dibenz- [b,f]azepin-5-propanamin 280,4 250.10-6 ICN Biomedica Is, Irvine, CA, USA A, F
    Quinidin (2-ethenyl- 4-azabicyclo [2.2.2] oct- 5-yl)- (6-methoxyquinolin- 4-yl)-methanol 324,4 100. 10-6 A, E
    Verapamil 5-[N-(3,4-dimethoxyphenylethyl)methy lamino]-2-(3,4-dimethoxyphenyl)-2-isopropylvaleronitrile hydrochloride 491.06 100.10-6 Sigma-Aldrich, St. Louis, MO USA D
    Niflumic acid 2-[3-(trifluoromethyl)anilino]nicoti nic acid, 2-(α,α,α-trifluoro-m-toluidino)nicotinic acid 282.22 100.10-6 Sigma-Aldrich, St. Louis, MO USA C
    H2DIDS 4,4'-diisothiocyanatodihydrostil-bene-2,2'-disulfonic acid, disodium salt 500.48 100.10-6 Molecular Probes, Eugene, OR USA B, C
    NBBP 5-nitro-2-(3-phenyl-propylamino)benzoic acid 300.31 100.10-6 Sigma-Aldrich, St. Louis, MO USA C
    A: inhibitor of Na+ channels and of Na+-dependent Mg2+ efflux system
    B: wide range chloride (anion) transporter inhibitor
    C: CI channel inhibitor
    D: L-type Ca2+ channels inhibitor
    E: K+ channel inhibitor
    F: Na+ channel inhibitor
  • Results [Mg2+]i of -tet and +tet HEK293-(SLC41A1) cells.
  • The basal [Mg2+]i of Mag-Fura 2-loaded -tet and +tet (15 hrs) HEK293-(SLC41A1) cells was measured in completely Mg-free but Na-containing medium after a 20-min pre-incubation in the same solution (table 7). Compared to -tet cells (0.26 ± 0.05 mM), tetracycline-induced HEK293-(SLC41A1) cells had a 62% lower [Mg2+]i (0.10 ± 0.02 mM). In agreement with our former results [Kolisek et al., 2008] these data point to an increased efflux capacity of HEK293 cells over-expressing SLC41A1. To proof this, the efflux capacity of differently Mg-loaded -tet and +tet cells was determined.
  • [Mg 2+ ]i change in differently Mg-loaded-tet and +tet HEK293-(SLC41A1) cells.
  • As it is well known that the [Mg2+]i influences the Mg2+ efflux activity, the experiments were designed to support SLC41A1-related Mg2+ release by performing all preparation and storage procedures (with exception of probe-loading) before the actual measurements in Mg-containing solutions. In addition, directly before the efflux experiments, -tet and +tet HEK293-(SLC41A1) cells were differently Mg-loaded by a 20-min incubation in media containing 2, 5 or 10 mmol.l-1 Mg. Afterwards, the Mg2+ efflux capacity was determined as [Mg2+]i decrease measured over a 20-mins period in completely Mg-free media. In table 6, [Mg2+]i values determined at the beginning and end of the measuring period are given for all conditions. It can be seen that -tet HEK293-(SLC41A1) cells had a stable [Mg2+]i of 0.25 ± 0.02 mM and no increase or decrease was observed after Mg-loading or incubation in completely Mg-free medium. In contrast, pre-incubation with increasing extracellular Mg2+ concentrations induced a ≈3- to 4-fold elevation of the [Mg2+]i in +tet HEK293-(SLC41A1) cells (table 7). When re-suspended in absolutely Mg2+-free media these cells show a continuous [Mg2+]i decrease (fig. 1A) reflecting Mg2+ release from the cells. Depending on loading conditions (2, 5 or 10 mM extracellular Mg) this Mg2+ efflux results in an 48%, 53% and 65% reduction of the endpoint [Mg2+]i compared to the respective starting level (Table 7). The results for -tet and +tet (15 hrs) HEK293-(SLC41A1) cells are summarized in fig. 1B. In comparison to non-induced -tet HEK293-(SLC41A1) cells, a significant Mg2+ extrusion occurred when +tet HEK293-(SLC41A1) cells pre-loaded with 5 or 10 mM Mg2+ were suspended in Mg-free medium. The [Mg2+]i decrease during the measuring period amounted to 133 ± 33, 224 ± 51 and 257 ± 44 µmol.l-1 per 20 mins after pre-loading with 2, 5 and 10 mM of Mg, respectively (fig. 1B). Table 7: [Mg2+]i (mmol.l-1) of non-induced (-tet) and induced (+tet) HEK293-(SLC41A1) cells measured after loading with various [Mg2+]e.
    [Mg2+]e -tet (control) +tet
    mM [Mg2+]¡ at start [Mg2+]i at end [Mg2+]¡ at start [Mg2+]¡ at end
    0 0.24 ± 0.07 0.28 ± 0.07 0.10 ± 0.02 0.10 ± 0.02A
    2 0.23 ± 0.06 0.20 ± 0.06 0.27 ± 0.06 0.14 ± 0.04
    5 0.28 ± 0.07 0.27 ± 0.05 0.43 ± 0.08** 0.20 ± 0.04a
    10 0.26 ± 0.05 0.27 ± 0.05 0.40 ± 0.04** 0.14 ± 0.02b
    [Mg2+]i values achieved after 20 min in loading solutions with various [Mg2+]e of 0, 2, 5 or 10 mM ([Mg2+]i at start) and after 20 min in complete Mg-free media ([Mg2+]i at end) are given. Data are presented as means ± SE of 8 to 12 single experiments. A P < 0.05 vs. control (-tet cells); ** P < 0.001 vs. non-loaded +tet HEK293-(SLC41A1) cells ([Mg2+]e = 0 mM); a P < 0.05 vs. control ([Mg2+]i of +tet cells at start), b < 0.001 vs. control ([Mg2+]i of +tet cells at start)
  • Until now, none of the postulated Na+- or anion-dependent Mg2+ efflux systems have been identified at a molecular level. Therefore, the next step was the performance of experiments to define the transport characteristics of SLC41A1-related Mg2+ extrusion.
  • Mg 2+ efflux from hSLC41A1 depends on extracellular Na+ and is inhibited by the unspecific Na +/Mg 2+ exchanger inhibitors imipramine and quinidine.
  • First, the effect of the extracellular [Na+] on the Mg2+ extrusion was tested with Mg-loaded (10 mM) -tet and +tet HEK293-(SLC41A1) cells. The latter were suspended in absolutely Mg-free media containing 145 mM Na+ or in the same medium but all Na+ iso-osmotically replaced by the organic non-permeant cation NMDG and the [Mg2+]i was measured over a 20-minute period (figure 2A). In -tet control cells a steady [Mg2+]i of 0.45 ± 0.03 mM was determined. In contrast, the [Mg2+]i of +tet cells incubated in Na-containing media decreased from 0.74 ± 0.07 to 0.29 ± 0.04 mM demonstrating SLC41A1-related Mg2+ extrusion (fig. 2B). This Mg2+ efflux was clearly dependent on extracellular Na+ (Fig. 2). It decreased from 358 ± 69 µM/20 min in 145-Na+ medium to 55 ± 26 µM/20 min in 0-Na+NMDG medium (fig. 2C) reflecting a 82 ± 5% reduction. These results clearly point to a Na-dependent mechanism of SLC41A1-related Mg2+ efflux.
  • Therefore, we next evaluated whether application of known unspecific inhibitors of the Na+/Mg2+ exchanger resulted in changes of SLC41A1-mediated Mg2+ efflux. Mg2+-loaded (20 min, 10 mM) +tet HEK293-(SLC41A1) cells were incubated in a completely Mg2+-free medium with an [Na+]e of 145 mM without or with 250 µM imipramine (Fig. 3A) or 100 µM quinidin (Fig. 3C), respectively, and the [Mg2+]i was measured for 20 min. To correct for molecular leakiness, control measurements were done with -tet cells. As shown in figure 3A, application of imipramine diminished the SLC41A1-related Mg2+ efflux. Data are summarized in figure 3B, which shows that inhibitor application reduced Mg2+ efflux from 341 ± 35 to 129 ± 32 µM/20min corresponding to 64 ± 9%. Figure 3C shows a summary of the inhibitory effect of 100 µM quinidin on SLC41A1-mediated Mg2+ efflux. It decreased from 327 ± 71 µM/20 min to 176 ± 0.5 µM/20 min corresponding to a 38 ± 7% reduction.
  • hSLC41A1-mediated Mg2+ efflux is stimulated by dB-cAMP and angiotensin II but inhibited by insulin.
  • Depending on the loading conditions and thus, on the intracellular Mg2+ status, application of dB-cAMP decreases (loading in 10 mM-Mg2+ solution) SLC41A1-related Mg2+ efflux by 17% or increases (loading in 2 mM- and 0 mM-Mg2+ loading solution) it by 13% and 36%, respectively (Figure 4). Interestingly, application of dB-cAMP to growing SLC41A1-overexpressing cells led to a stimulation of their proliferation (Figure 5). In contrast, the Na+/Mg2+ exchanger inhibitor imipramin reduced the proliferation of such cells compared to controls (Figure 5).
    In addition, angiotensin II stimulated SLC41A1-related Mg2+ efflux by 19,5%.
    As shown in figure 6, insulin reduced the SLC41A1-mediated Mg2+ efflux by 38% (200 µU7ml) and 68% (400 µU/ml), respectively.
  • The Ca 2+ channel inhibitor Verapamil has no effect on hSLC41A1-mediated Mg2+ efflux.
  • In cardiac myocytes Verapamil has been show to effect Na+-dependent Mg2+ efflux by an unknown mechanism (Handy et al., 1996). Thus we tested the effect of this L-type Ca2+ channel inhibitor on Mg2+ efflux from +tet HEK293-(SLC41A1) cells. Although we used relatively high concentrations of 100 µM and 200 µM, Verapamil, respectively, we found no effect of the inhibitor on SLC41A1-mediated Mg2+ extrusion. The observed [Mg2+]i decrease amounted to 344 ± 41 µM / 20 min under control conditions and to 386 ± 45 µM / 20 min or 326 ± 58 µM / 20 min after application of 100 µM or 200 µM of verapamil.
  • References
    1. 1. Feil R. (2007). Conditional somatic mutagenesis in the mouse using site-specific recombinases. In Conditional Mutagenesis: An Approach to Disease Models, eds. R. Feil and D. Metzger, Springer-Verlag, Handb Exp Pharmacol 178, 3-28.
    2. 2. Goytain A, Quamme GA. Identification and characterization of a novel mammalian Mg2+ transporter with channel-like properties. BMC Genomics 2005a; 6: 48.
    3. 3. Goytain A, Quamme, GA. Functional characterization of ACDP2 (ancient conserved domain protein), a divalent metal transporter. Physiol Genomics 2005b; 22: 382-89.
    4. 4. Goytain, A.; Quamme GA. Functional characterization of the human solute carrier, SLC41A2. Biochem Biophys Res Commun 2005c; 330: 701-5
    5. 5. Goytain A, Quamme GA. Functional characterization of human SLC41A1, a Mg2+ transporter with similarity to the procaryotic MgtE Mg2+ transporters. Physiol Genomics 2005d; 21: 337-42.
    6. 6. Goytain A, Hines, RM, El-Husseini A, Quamme GA. NIPA1 (SPG6), the basis for autosomal dominant form of hereditary spastic paraplegia, encodes a functional Mg2+ transporter. J Biol Chem 2007; 282: 8060-68.
    7. 7. GOnther T, Vormann J, Förster R (1984) Regulation of intracellular magnesium by Mg2+ efflux. Biochem Biophys Res Comm. 119: 124-131.
    8. 8. Handy RD, Gow IF, Ellis D, Flatman PW. Na-dependent regulation of intracellular free magnesium concentration in isolated rat ventricular myocytes. J Mol cell Cardiol 1996, 28: 1641-1651.
    9. 9. Kimura M (2007) Overview of magnesium nutrition. In: Nishizawa Y, Morii H, Durlach J (Eds.): New perspectives in magnesium research: nutrition and health. Springer, London, 2007, 69-93.
    10. 10. Kolisek M, Zsurka G, Samaj J, Weghuber J, Schweyen RJ, Schweigel, M. Mrs2p is an essential component of the major electrophoretic Mg2+ influx system in mitochondria. EMBO J 2003; 22: 1235-44.
    11. 11. Kolisek M, Launay P, Beck A, Sponder G, Serafini M, Brenkus M, Froschauer EM, Martens H, Fleig A, Schweigel M. (2008) SLC41A1 is a novel mammalian Mg2+ carrier. J Biol Chem; 283: 16235-47.
    12. 12. Kolisek M, Schweyen RJ, Schweigel M. (2007) Cellular Mg2+ transport and homeosasis: An overview. In: Nishizawa Y, Morii H, Durlach J (Eds.): New perspectives in magnesium research: nutrition and health. Springer, London, 2007, 21-33.
    13. 13. Kumeda Y, Inaba M. Review of magnesium and metabolic syndrome. In: Nishizawa Y, Morii H, Durlach J (Eds.): New perspectives in magnesium research: nutrition and health. Springer, London, 2007, 189-196.
    14. 14. Ödblom MP, Handy RD (1999) A novel DIDS-sensitive, anion-dependent Mg2+ efflux pathway in rat ventricular myocytes. Biochem. Biophys. Res. Comm. 264: 334-337.
    15. 15. Po SS; Wang DW, Yang IC, Johnson JP, Nie L, Bennett, PB. Modulation of HERG potassium channels by extracellular magnesium and quinidine. J Cardiovasc Pharmacol 1999, 33:181-185.
    16. 16. Quamme GA. (1989) Control of magnesium transport in the thick ascending limb. Am. J. Physiol. Renal Fluid Electrolyte Physiol. 25: F197-F210.
    17. 17. Resnick LM, Gupta RK, Laragh JH (1984) Intracellular free magnesium in erythrocytes of essential hypertension: relation to blood pressure and serum divalent cations. Proc Natl Acad Sci USA 81, 6511-6515.
    18. 18. Romanuik TL, Wang G, Holt RA; Jones SJM Marra MA, Sadar MD (2009) Identification of novel androgen-responsive genes by sequencing of LongSAGE libraries. BMC Genomics 10: 476-95.
    19. 19. Sahni J, Nelson B, Scharenberg AM. SLC41A2 encodes a plasma-membrane Mg2+ transporter. Biochem J 2007; 401: 505-13.
    20. 20. Schlingmann KP, Weber S, Peters M, Niemann-Nejsum L, Vitzhum H, Klingel K, Kratz M, Haddad E, Ristoff E, Dinour D, Syrrou M, Nielsen S, Sassen M, Waldegger S, Seyberth HW, Konrad M. Hypomagnesemia with secondary hypocalcemia is caused by mutations in TRPM6, a new member of the TRMP gene family. Nature Genetics 2002; 31: 166-70.
    21. 21. Schmitz C, Perraud AL, Johnson CO, Inabe K, Smith MK, Penner R, Kurosaki T, Fleig A, Scharenberg AM. Regulation of vertebrate cellular Mg2+-homeostasis by TRPM7. Cell 2003; 114: 191-200.
    22. 22. Schweigel M, Park H-S, Etschmann B, Martens H. Characterization of the Na+-dependent Mg2+ transport in sheep ruminal epithelial cells. Am J Physiol Gastrointest Liver Physiol 2006; 290: G56-G65.
    23. 23. Schweigel M, Park H-S, Etschmann B, Martens H. (2006) Characterization of the Na+-dependent Mg2+ transport in sheep ruminal epithelial cells. Am J Physiol Gastrointest Liver Physiol 290: G56-G65.
    24. 24. Smith, R.L., Thompson, L.J., and Maguire, M.E. (1995) J Bacteriol 177, 1233-8
    25. 25. Sponder G, Svidova S, Schweigel M, Vormann J, Kolisek M. (2010) Splice-variant 1 of the ancient domain protein 2 (ACDP2) complements the magnesium-deficient growth phenotype of Salmonella enferica sv. typhimurium strain MM281. Magnesium Research 23, 105-14.
    26. 26. Schweigel M, Vormann J, Martens H (2000) Mechanisms of Mg2+ transport in cultured rumen epithelial cells. Am J Physiol Gastrointest Liver Physiol 278: G400-G408.
    27. 27. Wabakken, T., Rian, E., Kveine, M., and Aasheim, H.C. (2003) Biochem Biophys Res Commun 306, 718-24
    28. 28. Watanabe M, Konishi M, Ohkido I, Matsufuji S. (2005) Enhanced sodium-dependent extrusion of magnesium in mutant cells established from a mouse renal tubular cell line. Am J Physiol Renal Physiol; 289, 742-48.
    29. 29. Zhou H, Clapham DE. (2009) Mammalian MagT1 and TUSC3 are required for cellular magnesium uptake and vertebrate embryonic development. Proc Natl Acad Sci U S A.: 106, 15750-5.
    30. 30. Johnsson and Varshavsky, Proc. Natl. Acad. Sci., 1994, 91 (22), 10340-10344, in U.S. patent 5,503,977 A or U.S. patent 5,585,245 A .
    31. 31. Shibahara Kei-ichi et al. in published US patent application 20100003686
    SEQUENCE LISTING
    • <110> FBN - Leibniz-Institut für Nutztierbiologie, Dummerstorf
    • <120> Na+/Mg2+ exchanger
    • <130> PVA-24 699 EP
    • <160> 5
    • <170> PatentIn version 3.3
    • <210> 1
      <211> 513
      <212> PRT
      <213> Homo sapiens
    • <400> 1
      Figure imgb0001
      Figure imgb0002
      Figure imgb0003
    • <210> 2
      <211> 1542
      <212> RNA
      <213> homo sapiens
    • <400> 2
      Figure imgb0004
      Figure imgb0005
    • <210> 3
      <211> 1542
      <212> DNA
      <213> homo sapiens
    • <400> 3
      Figure imgb0006
      Figure imgb0007
    • <210> 4
      <211> 1539
      <212> DNA
      <213> Mus musculus
    • <400> 4
      Figure imgb0008
      Figure imgb0009
    • <210> 5
      <211> 512
      <212> PRT
      <213> Mus musculus
    • <400> 5
      Figure imgb0010
      Figure imgb0011
      Figure imgb0012

Claims (1)

  1. Mutation library for the identification or generation of SLC41A1 variants, wherein the protein SLC41A1 having the amino acid sequence of SEQ ID NO 1 is the cellular Na+/Mg2+ exchanger, comprising
    i) proteins with mutations of SEQ ID NO 1, wherein one of the amino acids of SEQ ID NO 1 is substituted at positions 21 (S>N), 25 (S>Q), 73 (S>N), 76 (S>N), 80 (S>N), 89 (S>N), 97 (S>N), 158 (S>N), 309 (S>N), 394 (S>N), 408 (S>Q), 81 (T>Q), 168 (T>Q), 509 (T>Q) or 278 (Y>N) and proteins with at least one mutation of SEQ ID NO 1, wherein a plurality of these substitutions is present; wherein the mutation library consists of 15 peptides with a single aa mutation and at least one peptide wherein a plurality of these substitutions is present
    or
    ii) proteins with mutations of the transmembrane domains and N-, C- termini mutants of SEQ ID NO 1, wherein the amino acids 2-30, 31-60, 61-90, 2-60, 2-90 or 504-513 of SEQ ID NO 1 are deleted (6 mutated proteins)
    and
    proteins with mutations of SEQ ID NO 1 in membrane spanning domains, wherein the amino acids 96-205, 225-279, 286-337, 348-427 or 439-503 are deleted (5 mutated proteins)
    and proteins with mutations in membrane spanning domains of SEQ ID NO 1,
    wherein the amino acids 99-150, 180-241, 256-306, 317-368, 408-459 or 437-503 are deleted (6 mutated proteins); wherein the mutation library consists of 17 peptides
    or
    iii) proteins with leucine zipper (LZ) mutations of SEQ ID NO 1, wherein the amino acids 482-503 are deleted and wherein amino acids in LZ domain in positions 482 (L>V), 489 (L>V), 494 (L>V), 496 (L>V), 501 (L>V) or 503 (L>V) are substituted; wherein the mutation library consists of 7 peptides or
    iv) proteins with external loop (EL) mutations of SEQ ID NO 1, wherein the amino acids 128-179, 247-254, 310-315, 369-405, or 468-478 are deleted; wherein the mutation library consists of 5 peptides.
EP10177510.4A 2010-09-18 2010-09-18 Na+/Mg2+ exchanger Active EP2431386B1 (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
EP10177510.4A EP2431386B1 (en) 2010-09-18 2010-09-18 Na+/Mg2+ exchanger
PCT/EP2011/065979 WO2012035088A1 (en) 2010-09-18 2011-09-15 Na+/Mg2+ exchanger
JP2013528667A JP2013543486A (en) 2010-09-18 2011-09-15 Na + / Mg2 + exchanger
CA2811538A CA2811538C (en) 2010-09-18 2011-09-15 Na+/mg2+ exchanger
US13/823,383 US20130174289A1 (en) 2010-09-18 2011-09-15 Na+/Mg2+ Exchanger

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
EP10177510.4A EP2431386B1 (en) 2010-09-18 2010-09-18 Na+/Mg2+ exchanger

Publications (2)

Publication Number Publication Date
EP2431386A1 EP2431386A1 (en) 2012-03-21
EP2431386B1 true EP2431386B1 (en) 2015-02-25

Family

ID=43480818

Family Applications (1)

Application Number Title Priority Date Filing Date
EP10177510.4A Active EP2431386B1 (en) 2010-09-18 2010-09-18 Na+/Mg2+ exchanger

Country Status (5)

Country Link
US (1) US20130174289A1 (en)
EP (1) EP2431386B1 (en)
JP (1) JP2013543486A (en)
CA (1) CA2811538C (en)
WO (1) WO2012035088A1 (en)

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5585245A (en) 1994-04-22 1996-12-17 California Institute Of Technology Ubiquitin-based split protein sensor
US5503977A (en) 1994-04-22 1996-04-02 California Institute Of Technology Split ubiquitin protein sensor
US20020076750A1 (en) * 2000-05-25 2002-06-20 Ming-Hui Wei Isolated human transporter proteins, nucleic acid molecules encoding human transporter proteins, and uses thereof
DE602007012460D1 (en) 2006-10-23 2011-03-24 Res Org Information & Systems IGNITION FOR TETRACYCLIN-INDUCED GENE EXPRESSION OR A CELL STEM WITH CONDITIONAL GENE KNOCKOUT, AND USE OF CELL STRAINS

Also Published As

Publication number Publication date
US20130174289A1 (en) 2013-07-04
EP2431386A1 (en) 2012-03-21
CA2811538C (en) 2019-09-03
WO2012035088A1 (en) 2012-03-22
CA2811538A1 (en) 2012-03-22
JP2013543486A (en) 2013-12-05

Similar Documents

Publication Publication Date Title
Forrest et al. Targeted disruption of NMDA receptor 1 gene abolishes NMDA response and results in neonatal death
Stenvers et al. Heart and liver defects and reduced transforming growth factor β2 sensitivity in transforming growth factor β type III receptor-deficient embryos
Janz et al. SV2A and SV2B function as redundant Ca2+ regulators in neurotransmitter release
Link et al. Targeted inactivation of the gene encoding the mouse alpha 2c-adrenoceptor homolog.
Sun et al. Mosaic mutant analysis identifies PDGFRα/PDGFRβ as negative regulators of adipogenesis
Soloff et al. Targeted deletion of protein kinase C λ reveals a distribution of functions between the two atypical protein kinase C isoforms
JP5340242B2 (en) G protein-related mutants in essential hypertension
Schwindinger et al. Mice with deficiency of G protein γ3 are lean and have seizures
Surmacz et al. Dissociation of mitogenesis and transforming activity by C-terminal truncation of the insulin-like growth factor-I receptor
Hata et al. Expression and molecular pharmacology of the mouse CRTH2 receptor
JP5940541B2 (en) Neuropeptide Q as modulator of GPCR GALR2 and use thereof
Xiong et al. Cardiac-specific, inducible ClC-3 gene deletion eliminates native volume-sensitive chloride channels and produces myocardial hypertrophy in adult mice
US7718846B2 (en) Animal models of long QT syndrome and uses thereof
US6093873A (en) Genetically engineered mice containing alterations in the gene encoding RXR
Kaur et al. RGS-insensitive Gα subunits: probes of Gα subtype-selective signaling and physiological functions of RGS proteins
US20070135365A1 (en) Pharmaceutical composition for preventing or remedying cardiac hypertrophy and cardiovascular disease caused thereby
ES2432759T3 (en) New diagnostic methods
EP2431386B1 (en) Na+/Mg2+ exchanger
JP2011501672A (en) Novel therapeutic targets in bowel disease
WO2001058944A1 (en) Sodium-independent small neutral amino acid transporters transporting l- and d-amino acids and genes thereof
Zhu et al. Enhanced calcium cycling and contractile function in transgenic hearts expressing constitutively active Gαo* protein
JP2007505628A (en) Modulation of cardiac contractility and heart failure tendency
JP2013543978A (en) GPR101 transgenic mouse, its use and screening method
Kaura Mechanisms underlying the phenotypic diversity in RYR1-associated Malignant Hyperthermia
Sun et al. Overexpressing of the GIPC1 protects against pathological cardiac remodelling

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME RS

17P Request for examination filed

Effective date: 20120917

RIN1 Information on inventor provided before grant (corrected)

Inventor name: ROENTGEN, MONIKA

Inventor name: KOLISEK, MARTIN

17Q First examination report despatched

Effective date: 20130918

RIC1 Information provided on ipc code assigned before grant

Ipc: C12N 15/12 20060101ALI20140703BHEP

Ipc: C07K 14/705 20060101AFI20140703BHEP

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

INTG Intention to grant announced

Effective date: 20140904

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO SE SI SK SM TR

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602010022453

Country of ref document: DE

Effective date: 20150409

REG Reference to a national code

Ref country code: AT

Ref legal event code: REF

Ref document number: 711918

Country of ref document: AT

Kind code of ref document: T

Effective date: 20150415

REG Reference to a national code

Ref country code: CH

Ref legal event code: NV

Representative=s name: ANDRE ROLAND S.A., CH

REG Reference to a national code

Ref country code: SE

Ref legal event code: TRGR

REG Reference to a national code

Ref country code: NL

Ref legal event code: VDEP

Effective date: 20150225

REG Reference to a national code

Ref country code: AT

Ref legal event code: MK05

Ref document number: 711918

Country of ref document: AT

Kind code of ref document: T

Effective date: 20150225

REG Reference to a national code

Ref country code: LT

Ref legal event code: MG4D

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150225

Ref country code: HR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150225

Ref country code: FI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150225

Ref country code: NO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150525

Ref country code: ES

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150225

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150625

Ref country code: AT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150225

Ref country code: LV

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150225

Ref country code: GR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150526

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: NL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150225

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: EE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150225

Ref country code: RO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150225

Ref country code: CZ

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150225

Ref country code: DK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150225

Ref country code: SK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150225

REG Reference to a national code

Ref country code: DE

Ref legal event code: R097

Ref document number: 602010022453

Country of ref document: DE

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: PL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150225

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150225

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

26N No opposition filed

Effective date: 20151126

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150225

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LU

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150918

Ref country code: MC

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150225

REG Reference to a national code

Ref country code: IE

Ref legal event code: MM4A

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20150918

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 7

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150225

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: BG

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150225

Ref country code: SM

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150225

Ref country code: HU

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT; INVALID AB INITIO

Effective date: 20100918

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: CY

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150225

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: TR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150225

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 8

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: PT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150225

Ref country code: MK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150225

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 9

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: AL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150225

REG Reference to a national code

Ref country code: DE

Ref legal event code: R082

Ref document number: 602010022453

Country of ref document: DE

Representative=s name: ETL IP PATENTANWALTSGESELLSCHAFT MBH, DE

Ref country code: DE

Ref legal event code: R082

Ref document number: 602010022453

Country of ref document: DE

Representative=s name: ETL IP PATENT- UND RECHTSANWALTSGESELLSCHAFT M, DE

Ref country code: DE

Ref legal event code: R082

Ref document number: 602010022453

Country of ref document: DE

Representative=s name: ETL WABLAT & KOLLEGEN PATENT- UND RECHTSANWALT, DE

REG Reference to a national code

Ref country code: DE

Ref legal event code: R082

Ref document number: 602010022453

Country of ref document: DE

Representative=s name: ETL IP PATENTANWALTSGESELLSCHAFT MBH, DE

Ref country code: DE

Ref legal event code: R082

Ref document number: 602010022453

Country of ref document: DE

Representative=s name: ETL IP PATENT- UND RECHTSANWALTSGESELLSCHAFT M, DE

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: CH

Payment date: 20210923

Year of fee payment: 12

Ref country code: FR

Payment date: 20210927

Year of fee payment: 12

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: BE

Payment date: 20210921

Year of fee payment: 12

Ref country code: SE

Payment date: 20210921

Year of fee payment: 12

Ref country code: GB

Payment date: 20210923

Year of fee payment: 12

REG Reference to a national code

Ref country code: CH

Ref legal event code: PL

REG Reference to a national code

Ref country code: SE

Ref legal event code: EUG

GBPC Gb: european patent ceased through non-payment of renewal fee

Effective date: 20220918

REG Reference to a national code

Ref country code: BE

Ref legal event code: MM

Effective date: 20220930

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LI

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20220930

Ref country code: FR

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20220930

Ref country code: CH

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20220930

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20220919

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: BE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20220930

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: GB

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20220918

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: DE

Payment date: 20230901

Year of fee payment: 14